1
|
Patel VI, Metcalf JP. Airway Macrophage and Dendritic Cell Subsets in the Resting Human Lung. Crit Rev Immunol 2019; 38:303-331. [PMID: 30806245 DOI: 10.1615/critrevimmunol.2018026459] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Dendritic cells (DCs) and macrophages (MΦs) are antigen-presenting phagocytic cells found in many peripheral tissues of the human body, including the blood, lymph nodes, skin, and lung. They are vital to maintaining steady-state respiration in the human lung based on their ability to clear airways while also directing tolerogenic or inflammatory responses based on specific stimuli. Over the past three decades, studies have determined that there are multiple subsets of these two general cell types that exist in the airways and interstitium. Identifying these numerous subsets has proven challenging, especially with the unique microenvironments present in the lung. Cells found in the vasculature are not the same subsets found in the skin or the lung, as demonstrated by surface marker expression. By transcriptional profiling, these subsets show similarities but also major differences. Primary human lung cells and/ or tissues are difficult to acquire, particularly in a healthy condition. Additionally, surface marker screening and transcriptional profiling are continually identifying new DC and MΦ subsets. While the overall field is moving forward, we emphasize that more attention needs to focus on replicating the steady-state microenvironment of the lung to reveal the physiological functions of these subsets.
Collapse
Affiliation(s)
- Vineet Indrajit Patel
- Pulmonary and Critical Care Division of the Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Jordan Patrick Metcalf
- Pulmonary and Critical Care Division of the Department of Medicine and Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
2
|
Noto FK, Adjan-Steffey V, Tong M, Ravichandran K, Zhang W, Arey A, McClain CB, Ostertag E, Mazhar S, Sangodkar J, DiFeo A, Crawford J, Narla G, Jamling TY. Sprague Dawley Rag2-Null Rats Created from Engineered Spermatogonial Stem Cells Are Immunodeficient and Permissive to Human Xenografts. Mol Cancer Ther 2018; 17:2481-2489. [PMID: 30206106 DOI: 10.1158/1535-7163.mct-18-0156] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 07/10/2018] [Accepted: 09/05/2018] [Indexed: 12/22/2022]
Abstract
The rat is the preferred model for toxicology studies, and it offers distinctive advantages over the mouse as a preclinical research model including larger sample size collection, lower rates of drug clearance, and relative ease of surgical manipulation. An immunodeficient rat would allow for larger tumor size development, prolonged dosing and drug efficacy studies, and preliminary toxicologic testing and pharmacokinetic/pharmacodynamic studies in the same model animal. Here, we created an immunodeficient rat with a functional deletion of the Recombination Activating Gene 2 (Rag2) gene, using genetically modified spermatogonial stem cells (SSC). We targeted the Rag2 gene in rat SSCs with TALENs and transplanted these Rag2-deficient SSCs into sterile recipients. Offspring were genotyped, and a founder with a 27 bp deletion mutation was identified and bred to homozygosity to produce the Sprague-Dawley Rag2 - Rag2 tm1Hera (SDR) knockout rat. We demonstrated that SDR rat lacks mature B and T cells. Furthermore, the SDR rat model was permissive to growth of human glioblastoma cell line subcutaneously resulting in successful growth of tumors. In addition, a human KRAS-mutant non-small cell lung cancer cell line (H358), a patient-derived high-grade serous ovarian cancer cell line (OV81), and a patient-derived recurrent endometrial cancer cell line (OV185) were transplanted subcutaneously to test the ability of the SDR rat to accommodate human xenografts from multiple tissue types. All human cancer cell lines showed efficient tumor uptake and growth kinetics indicating that the SDR rat is a viable host for a range of xenograft studies. Mol Cancer Ther; 17(11); 2481-9. ©2018 AACR.
Collapse
Affiliation(s)
| | | | - Min Tong
- Poseida Therapeutics Inc., San Diego, California
| | | | - Wei Zhang
- Hera BioLabs Inc., Lexington, Kentucky
| | | | | | - Eric Ostertag
- Transposagen Biopharmaceuticals Inc., Lexington, Kentucky
| | - Sahar Mazhar
- Case Western Reserve University, Cleveland, Ohio
| | | | | | - Jack Crawford
- Hera BioLabs Inc., Lexington, Kentucky.,Transposagen Biopharmaceuticals Inc., Lexington, Kentucky
| | - Goutham Narla
- Hera BioLabs Inc., Lexington, Kentucky.,The University of Michigan, Ann Arbor, Michigan
| | - Tseten Y Jamling
- Hera BioLabs Inc., Lexington, Kentucky. .,Transposagen Biopharmaceuticals Inc., Lexington, Kentucky
| |
Collapse
|
3
|
Thiel A, Kesselring R, Pries R, Puzik A, Wittkopf N, Wollenberg B. Expression of the T cell receptor αβ on a CD123+ BDCA2+ HLA-DR+ subpopulation in head and neck squamous cell carcinoma. PLoS One 2011; 6:e15997. [PMID: 21264308 PMCID: PMC3019173 DOI: 10.1371/journal.pone.0015997] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Accepted: 12/02/2010] [Indexed: 11/19/2022] Open
Abstract
Human Plasmacytoid Dendritic Cells (PDCs) infiltrating solid tumor tissues and draining lymph nodes of Head and Neck Squamous Cell Carcinoma (HNSCC) show an impaired immune response. In addition to an attenuated secretion of IFN-α little is known about other HNSCC-induced functional alterations in PDCs. Particular objectives in this project were to gain new insights regarding tumor-induced phenotypical and functional alterations in the PDC population. We showed by FACS analysis and RT-PCR that HNSCC orchestrates an as yet unknown subpopulation exhibiting functional autonomy in-vitro and in-vivo besides bearing phenotypical resemblance to PDCs and T cells. A subset, positive for the PDC markers CD123, BDCA-2, HLA-DR and the T cell receptor αβ (TCR-αβ) was significantly induced subsequent to stimulation with HNSCC in-vitro (p = 0.009) and also present in metastatic lymph nodes in-vivo. This subgroup could be functionally distinguished due to an enhanced production of IL-2 (p = 0.02), IL-6 (p = 0.0007) and TGF-β (not significant). Furthermore, after exposure to HNSCC cells, mRNA levels revealed a D-J-beta rearrangement of the TCR-beta chain besides a strong enhancement of the CD3ε chain in the PDC population. Our data indicate an interface between the PDC and T cell lineage. These findings will improve our understanding of phenotypical and functional intricacies concerning the very heterogeneous PDC population in-vivo.
Collapse
Affiliation(s)
- Annette Thiel
- Department of Otorhinolaryngology, University of Schleswig-Holstein Campus Lübeck, Lübeck, Germany.
| | | | | | | | | | | |
Collapse
|
4
|
Chapiro E, Delabesse E, Asnafi V, Millien C, Davi F, Nugent E, Beldjord K, Haferlach T, Grimwade D, Macintyre EA. Expression of T-lineage-affiliated transcripts and TCR rearrangements in acute promyelocytic leukemia: implications for the cellular target of t(15;17). Blood 2006; 108:3484-93. [PMID: 16857994 DOI: 10.1182/blood-2005-09-009977] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Acute promyelocytic leukemia (APL) is the most differentiated form of acute myeloid leukemia (AML) and has generally been considered to result from transformation of a committed myeloid progenitor. Paradoxically, APL has long been known to express the T-cell lymphoid marker, CD2. We searched for other parameters indicative of T-cell lymphoid specification in a cohort of 36 APL cases, revealing a frequent but asynchronous T-cell lymphoid program most marked in the hypogranular variant (M3v) subtype, with expression of PTCRA, sterile TCRA, and TCRG transcripts and TCRG rearrangement in association with sporadic cytoplasmic expression of CD3 or TdT proteins. Gene-expression profiling identified differentially expressed transcription factors that have been implicated in lymphopoiesis. These data carry implications for the hematopoietic progenitor targeted by the PML-RARA oncoprotein in APL and are suggestive of a different cellular origin for classic hypergranular (M3) and variant forms of the disease. They are also consistent with the existence and subsequent transformation of progenitor populations with lymphoid/myeloid potential.
Collapse
MESH Headings
- Cell Lineage
- Cell Transformation, Neoplastic
- Chromosomes, Human, Pair 15
- Chromosomes, Human, Pair 17
- Cohort Studies
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic
- Gene Rearrangement, T-Lymphocyte
- Humans
- Leukemia, Promyelocytic, Acute/genetics
- Lymphopoiesis/genetics
- RNA, Messenger
- T-Lymphocytes/metabolism
- T-Lymphocytes/pathology
- Transcription Factors/genetics
- Translocation, Genetic
Collapse
Affiliation(s)
- Elise Chapiro
- Department of Hematology, Université Paris-Descartes, Faculté de Médecine, and Institut National de la Santé et de la Recherche Médicale (INSERM) EMI0210, Assistance Publique-Hôpitaux de Paris (AP-HP) Necker-Enfants Malades, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Mocchegiani E, Costarelli L, Giacconi R, Cipriano C, Muti E, Malavolta M. Zinc-binding proteins (metallothionein and alpha-2 macroglobulin) and immunosenescence. Exp Gerontol 2006; 41:1094-107. [PMID: 17030107 DOI: 10.1016/j.exger.2006.08.010] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2006] [Revised: 08/25/2006] [Accepted: 08/29/2006] [Indexed: 11/28/2022]
Abstract
Zinc is a relevant trace element for the efficiency of the entire immune system. The binding of zinc with some proteins, such as metallothioneins (MT) and alpha-2 macroglobulin (alpha-2M) is crucial for the immune efficiency during ageing and in age-related diseases, because these proteins may be involved in antagonistic pleiotropic effects. Indeed, the presence of chronic inflammation during ageing, generally, induces overexpression of these proteins that, due to their original biological function in fighting stressor agents, continuously sequester intracellular zinc. As a consequence, a low zinc ion availability may appear in aged organisms leading to impairments of the immune response at thymic and extrathymic levels with the risk of the appearance of age-related diseases. Therefore, MT and alpha-2M turn from protective in "young-adult age" to harmful agents in "ageing" following the basic assumption of an evolutionary theory of ageing, named the "antagonistic pleiotropy", which suggests that a trade off between early beneficial effects and late negative outcomes can occur at a genetic and molecular level. On the other hand, some polymorphisms of MT (MT2A) and alpha-2M have been associated with atherosclerosis or Alzheimer disease, respectively. Physiological zinc supplementation in elderly restores the thymic endocrine activity and innate immune response (NK cell cytotoxicity) and increases the survival rate in old mice. Therefore, zinc supplementation is useful to achieve health longevity because these zinc-binding proteins may regain their original protective task against oxidative damage with, thus, a beneficial impact on immune response.
Collapse
Affiliation(s)
- Eugenio Mocchegiani
- Immunology Center, Section: Nutrition, Immunity and Ageing, Research Department I.N.R.C.A., via Birarelli 8, 60121 Ancona, Italy.
| | | | | | | | | | | |
Collapse
|
6
|
Ito T, Liu YJ, Kadowaki N. Functional Diversity and Plasticity of Human Dendritic Cell Subsets. Int J Hematol 2005; 81:188-96. [PMID: 15814329 DOI: 10.1532/ijh97.05012] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The induction of different types of innate and adaptive immune responses, depending on the nature of the antigens and the environmental context, is crucial to cope with a variety of pathogens and concurrently to avoid pathologic reaction to self antigens. Recent studies have elucidated that the diversity of immune responses is critically controlled by dendritic cells (DCs). Two DC subsets, myeloid DCs and plasmacytoid DCs, have been identified in humans. The DC subsets recognize different microbial pathogens by expressing distinct repertoires of Toll-like receptors and induce different types of innate and adaptive immune responses, depending on the environmental factors. In particular, plasmacytoid DC precursors produce vast amounts of type I interferons in response to viruses and thus play an important role in antiviral immunity. Elucidating the cellular and molecular mechanisms that modulate the functions of the 2 DC subsets will lead to an understanding of the pathogenesis of various immune-related diseases and to the development of novel immunologic therapies.
Collapse
Affiliation(s)
- Tomoki Ito
- First Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | | | | |
Collapse
|
7
|
Arpinati M, Chirumbolo G, Urbini B, Bonifazi F, Bandini G, Saunthararajah Y, Zagnoli A, Stanzani M, Falcioni S, Perrone G, Tura S, Baccarani M, Rondelli D. Acute graft-versus-host disease and steroid treatment impair CD11c+ and CD123+ dendritic cell reconstitution after allogeneic peripheral blood stem cell transplantation. Biol Blood Marrow Transplant 2004; 10:106-15. [PMID: 14750076 DOI: 10.1016/j.bbmt.2003.09.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Human dendritic cells (DC) comprise 2 subsets-plasmacytoid CD123(+) and myeloid CD11c(+) DC-that may have distinct roles in the regulation of immunity after allogeneic hematopoietic stem cell transplantation. In this study, we analyzed the kinetics of CD123(+) DC and CD11c(+) DC reconstitution in 31 patients who underwent transplantation with allogeneic granulocyte colony-stimulating factor-mobilized peripheral blood (PB) stem cells from HLA-identical sibling donors after myeloablative conditioning. Lineage marker-negative HLA-DR(+) CD11c(+) CD11c(+) DC and lineage marker-negative HLA-DR(+) CD123(+) CD123(+) DC, as well as monocytes and lymphoid subsets, were enumerated in donor grafts and in the PB of patients at various time points after transplantation. Reconstitution of both CD11c(+) DC and CD123(+) DC to normal levels occurred within 6 to 12 months and was not affected by the diagnosis, preparatory regimen, or graft composition. However, PB CD11c(+) DC and CD123(+) DC counts were significantly reduced in patients with acute GVHD grade II to IV (at 1 and 3 months) and grade I (at 1 month). Patients with chronic GVHD instead showed reduced CD123(+) DC counts only 6 months after transplantation. Moreover, treatment with steroids (>0.1 mg/kg) was significantly associated with reduced PB CD11c(+) DC and CD123(+) DC counts at all time points after transplantation. In multivariate analysis, only acute GVHD affected DC reconstitution early after transplantation. These results will prompt new studies addressing whether DC reconstitution correlates with immunity against infectious agents or with graft-versus-tumor reactions after PB stem cell allotransplantation.
Collapse
Affiliation(s)
- Mario Arpinati
- Research Center for Transplant Immunology, Institute of Hematology and Medical Oncology Seràgnoli, University of Bologna, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Fohrer H, Audit IM, Sainz A, Schmitt C, Dezutter-Dambuyant C, Dalloul AH. Analysis of transcription factors in thymic and CD34+ progenitor-derived plasmacytoid and myeloid dendritic cells: evidence for distinct expression profiles. Exp Hematol 2004; 32:104-12. [PMID: 14725907 DOI: 10.1016/j.exphem.2003.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVE The expression of mRNA for pre-Talpha is specific for human plasmacytoid dendritic cells (PDC), a population ontogenically close to T cells. The latter need Gata-3 transcription factor to develop. PU1 and RelB are two transcription factors involved in the development of murine myeloid DC (MDC). To determine the lineage origin of human thymic DC, the expression of these genes was investigated. MATERIALS AND METHODS Fresh thymic DC, CD34(+)CD1a(-) progenitors, and progenitor-derived DC populations were sorted, analyzed, and compared to blood DC. RESULTS Three DC populations were found in the thymus. 1) CD123(-/lo)HLA-DR(hi) DC expressing PU1 and RelB; 2) CD123(hi)HLA-DR(+) DC expressing only pre-Talpha, the expression of which was similar to that of MDC and PDC from peripheral blood; and 3) a new mature CD123(hi)HLA-DR(hi) PDC population with pre-Talpha, PU1 and RelB mRNAs. In culture, most CD34(+)CD1a(-) progenitors remained CD1a(-)CD123(-); had a T and natural killer cell differentiation potential; and expressed Gata-3 mRNA contrary to DC precursors. A few cells (10%) became CD1a(+)CD123(+) expressing pre-Talpha, PU1, and RelB mRNAs and were able to differentiate into typical Langerhans cells with transforming growth factor-beta. Coculture of thymic progenitors on a murine cell line generated CD123(hi)CD1a(-) cells with typical PDC morphology, expressing pre-Talpha but not PU1 and RelB transcripts. Activated PDC acquired myeloid antigens, and up-regulated PU1 and RelB mRNAs while down-regulating pre-Talpha mRNA expression. CONCLUSION Both DC maturation pathways may arise from distinct precursors but are interconnected. DC differentiation seems to occur from Gata-3(-) precursors upstream of T and natural killer precursors.
Collapse
Affiliation(s)
- Hélène Fohrer
- INSERM U543, Hôpital Pitié-Salpêtrière, Paris, France
| | | | | | | | | | | |
Collapse
|
9
|
Bas A, Hammarström SG, Hammarström MLKC. Extrathymic TCR Gene Rearrangement in Human Small Intestine: Identification of New Splice Forms of Recombination Activating Gene-1 mRNA with Selective Tissue Expression. THE JOURNAL OF IMMUNOLOGY 2003; 171:3359-71. [PMID: 14500629 DOI: 10.4049/jimmunol.171.7.3359] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Two new 5'-untranslated region (5'UTR) exons were identified in the human gene for the lymphocyte-specific endonuclease recombination activating gene-1 (RAG1) required for the somatic recombination yielding functional Ag receptors. These 5'UTR exons were used in three different splice forms by jejunal lymphocytes of the T cell lineage. RAG1 mRNA containing the previously described 5'UTR exon was not expressed in these cells. Conversely, one of the new 5'UTR exons was not expressed in thymus. The new RAG1 mRNA splice forms were all expressed in immature T cells (CD2(+)CD7(+)CD3(-)). This cell population also expressed high levels of mRNA for the pre-T alpha-chain. In situ hybridization demonstrated jejunal cells expressing the new splice forms of RAG1 mRNA, both intraepithelially and in lamina propria. Pre-T alpha-chain mRNA-expressing cells were detected at the same sites. These results strongly suggest ongoing TCR gene rearrangement in human small intestinal mucosa, yielding T cells specially adapted for this environment. This seems to be achieved by two parallel processes, extrathymic T cell development and peripheral Ag-driven TCR editing.
Collapse
MESH Headings
- 5' Untranslated Regions/genetics
- 5' Untranslated Regions/immunology
- Adult
- Aged
- Base Sequence
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Child
- Exons/genetics
- Exons/immunology
- Female
- Gene Rearrangement, T-Lymphocyte
- Genes, RAG-1/immunology
- Humans
- Immunophenotyping
- Intestinal Mucosa/cytology
- Intestinal Mucosa/immunology
- Intestinal Mucosa/metabolism
- Jejunum/cytology
- Jejunum/immunology
- Jejunum/metabolism
- Jurkat Cells
- Lymphoid Tissue/cytology
- Lymphoid Tissue/immunology
- Lymphoid Tissue/metabolism
- Male
- Membrane Glycoproteins/biosynthesis
- Membrane Glycoproteins/blood
- Membrane Glycoproteins/genetics
- Middle Aged
- Molecular Sequence Data
- Organ Specificity/genetics
- Organ Specificity/immunology
- RNA Splicing/immunology
- RNA, Messenger/biosynthesis
- RNA, Messenger/isolation & purification
- RNA, Messenger/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/biosynthesis
- Receptors, Antigen, T-Cell, alpha-beta/blood
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Thymus Gland/cytology
- Thymus Gland/immunology
- Thymus Gland/metabolism
Collapse
Affiliation(s)
- Anna Bas
- Department of Clinical Microbiology, Division for Immunology, Umeå University, Umeå, Sweden
| | | | | |
Collapse
|
10
|
Arpinati M, Chirumbolo G, Urbini B, Perrone G, Rondelli D, Anasetti C. Role of plasmacytoid dendritic cells in immunity and tolerance after allogeneic hematopoietic stem cell transplantation. Transpl Immunol 2003; 11:345-56. [PMID: 12967787 DOI: 10.1016/s0966-3274(03)00055-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Dendritic cells (DC) may play an important role in the pathogenesis of alloimmune reactions, such as graft-vs.-host disease after allogeneic hematopoietic stem cell transplantation (HSCT). In humans, two types of DC-myeloid DC (mDC) and plasmacytoid DC (pDC) have been characterized and have distinct origins and functions. The data obtained from studies in vitro suggest that pDC are involved in the regulation of immunity, including the induction and maintenance of tolerance, as well as in the defence against viruses. The authors will review all the evidence currently available from reports exploring the role of pDC in clinical allogeneic HSCT.
Collapse
Affiliation(s)
- Mario Arpinati
- Centro di Ricerca sull'Immunologia dei Trapianti, Istituto di Ematologia e Oncologia Medica Seragnoli, Universita' di Bologna, Via Massarenti 9, Bologna 40138, Italy.
| | | | | | | | | | | |
Collapse
|
11
|
Sanz E, Alvarez-Mon M, Martínez-A C, de la Hera A. Human cord blood CD34+Pax-5+ B-cell progenitors: single-cell analyses of their gene expression profiles. Blood 2003; 101:3424-30. [PMID: 12446447 DOI: 10.1182/blood-2002-07-2244] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Circulating CD34(+) cells are used in reparative medicine as a stem cell source, but they contain cells already committed to different lineages. Many think that B-cell progenitors (BCPs) are confined to bone marrow (BM) niches until they differentiate into B cells and that they do not circulate in blood. The prevailing convention is that BCP transit a CD34(+)CD19(-)10(+) early-B-->CD34(+)CD19(+)CD10(+) B-cell progenitor (pro-B)-->CD34(-)CD19(+)CD10(+) B-cell precursor (pre-B) differentiation pathway within BM. However, populations of CD34(+)CD10(+) and CD34(+)CD19(+) cells circulate in adult peripheral blood and neonatal umbilical cord blood (CB) that are operationally taken as BCPs on the basis of their phenotypes, although they have not been submitted to a systematic characterization of their gene expression profiles. Here, conventional CD34(+)CD19(+)CD10(+) and novel CD34(+)CD19(+)CD10(-) BCP populations are characterized in CB by single-cell sorting and multiplex analyses of gene expression patterns. Circulating BCP are Pax-5(+) cells that span the early-B, pro-B, and pre-B developmental stages, defined by the profiles of rearranged V-D-J(H), CD79, VpreB, recombination activating gene (RAG), and terminal deoxynucleotidyl transferase (TdT) expression. Contrary to the expectation, circulating CD34(+)CD19(-)CD10(+) cells are essentially devoid of Pax-5(+) BCP. Interestingly, the novel CD34(+)CD19(+)CD10(-) BCP appears to be the normal counterpart of circulating preleukemic BCPs that undergo chromosomal translocations in utero months or years before their promotion into infant acute lymphoblastic B-cell leukemia after secondary postnatal mutations. The results underscore the power of single-cell analyses to characterize the gene expression profiles in a minor population of rare cells, which has broad implications in biomedicine.
Collapse
Affiliation(s)
- Eva Sanz
- Laboratory of Immunology and Oncology, Consejo Superior de Investigaciones Cientificas-Alcalá University Research Associated Unit, Madrid, Spain
| | | | | | | |
Collapse
|
12
|
Schotte R, Rissoan MC, Bendriss-Vermare N, Bridon JM, Duhen T, Weijer K, Brière F, Spits H. The transcription factor Spi-B is expressed in plasmacytoid DC precursors and inhibits T-, B-, and NK-cell development. Blood 2003; 101:1015-23. [PMID: 12393575 DOI: 10.1182/blood-2002-02-0438] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Human plasmacytoid dendritic cells (pDCs), also called type 2 dendritic cell precursors or natural interferon (IFN)-producing cells, represent a cell type with distinctive phenotypic and functional features. They are present in the thymus and probably share a common precursor with T and natural killer (NK) cells. In an effort to identify genes that control pDC development we searched for genes of which the expression is restricted to human pDC using a cDNA subtraction technique with activated monocyte-derived DCs (Mo-DCs) as competitor. We identified the transcription factor Spi-B to be expressed in pDCs but not in Mo-DCs. Spi-B expression in pDCs was maintained on in vitro maturation of pDCs. Spi-B was expressed in early CD34(+)CD38(-) hematopoietic progenitors and in CD34(+)CD1a(-) thymic precursors. Spi-B expression is down-regulated when uncommitted CD34(+)CD1a(-) thymic precursors differentiate into committed CD34(+)CD1a(+) pre-T cells. Overexpression of Spi-B in hematopoietic progenitor cells resulted in inhibition of development of T cells both in vitro and in vivo. In addition, development of progenitor cells into B and NK cells in vitro was also inhibited by Spi-B overexpression. Our results indicate that Spi-B is involved in the control of pDC development by limiting the capacity of progenitor cells to develop into other lymphoid lineages.
Collapse
Affiliation(s)
- Remko Schotte
- Division of Immunology of the Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Rossi M, Arpinati M, Rondelli D, Anasetti C. Plasmacytoid dendritic cells: do they have a role in immune responses after hematopoietic cell transplantation? Hum Immunol 2002; 63:1194-200. [PMID: 12480263 DOI: 10.1016/s0198-8859(02)00758-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Dendritic cells (DC) are the most potent antigen-presenting cells (APC) and are able to modulate immune responses. Investigators are studying methods to exploit the immunogenic and tolerogenic properties of DC. In the context of hematopoietic cell transplantation, DC might be helpful to facilitate engraftment and prevent graft-versus-host disease (GVHD) reactions. In this paper, we review circumstantial evidence that immature plasmacytoid DC might affect immune responses after transplantation of hematopoietic cells from allogeneic donors.
Collapse
Affiliation(s)
- Marco Rossi
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Department of Medicine, Division of Oncology, University of Washington, Seattle, Washington, USA
| | | | | | | |
Collapse
|
14
|
Ferrero I, Held W, Wilson A, Tacchini-Cottier F, Radtke F, MacDonald HR. Mouse CD11c(+) B220(+) Gr1(+) plasmacytoid dendritic cells develop independently of the T-cell lineage. Blood 2002; 100:2852-7. [PMID: 12351395 DOI: 10.1182/blood-2002-01-0214] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The developmental origin of dendritic cells (DCs) is controversial. In the mouse CD8alpha(+) and CD8alpha(-) DC subsets are often considered to be of lymphoid and myeloid origin respectively, although evidence on this point is conflicting. Very recently a novel CD11c(+) B220(+) DC subset has been identified that appears to be the murine counterpart to interferon alpha (IFNalpha)-producing human plasmacytoid DCs (PDCs). We show here that CD11c(+) B220(+) mouse PDCs, like human PDCs, are present in the thymus and express T lineage markers such as CD8alpha and CD4. However, the intrathymic development of PDCs can be completely dissociated from immature T lineage cells in mixed chimeras established with bone marrow cells from mice deficient for either Notch-1 or T-cell factor 1, two independent mutations that severely block early T-cell development. Our data indicate that thymic PDCs do not arise from a bipotential T/DC precursor.
Collapse
Affiliation(s)
- Isabel Ferrero
- Ludwig Institute for Cancer Research, Lausanne Branch, and the World Health Organization (WHO) Immunology Research and Training Center, Institute of Biochemistry, University of Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
15
|
Clark FJ, Chakraverty R. Role of dendritic cells in graft-versus-host disease. JOURNAL OF HEMATOTHERAPY & STEM CELL RESEARCH 2002; 11:601-16. [PMID: 12201949 DOI: 10.1089/15258160260194758] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
A major barrier to successful allogeneic hematopoietic stem cell transplantation is graft-versus-host disease (GVHD). Until recently, the role of antigen presentation in the development of this disorder was unknown. The experimental finding that recipient antigen-presenting cells (APCs) were required for the development of CD8(+) T cell-dependent GVHD has led to a fundamental reappraisal of our ideas concerning the pathogenesis of this disease. Following transplantation, the origin (donor or recipient), number, lineage, and function of APCs within the recipient are altered significantly. Studies that test the influence of each of these factors upon graft-versus-host responses, including graft-versus-tumor responses, are beginning to emerge and suggest that APCs, such as dendritic cells, constitute a potential target for therapeutic manipulation.
Collapse
Affiliation(s)
- Fiona J Clark
- Department of Haematology, Institute of Cancer Studies, University of Birmingham, Birmingham, UK
| | | |
Collapse
|
16
|
Comeau MR, Van der Vuurst de Vries AR, Maliszewski CR, Galibert L. CD123bright plasmacytoid predendritic cells: progenitors undergoing cell fate conversion? JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:75-83. [PMID: 12077231 DOI: 10.4049/jimmunol.169.1.75] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
CD123(bright) plasmacytoid cells (PC) and CD1c(+) peripheral blood myeloid dendritic cells (DC) are two human DC precursors that can be expanded in vivo by Fms-like tyrosine kinase 3 ligand (FL). It has been proposed that PC and myeloid CD1c(+) DC may represent two distinct lineages of DC. However, the phylogenetic affiliation of PC and its relationship with myeloid DC remain controversial. Here we show that CD123(bright)HLA-DR(+) PC from FL-treated healthy volunteers can be divided into mutually exclusive subsets that harbor either lymphoid or myeloid features. Lymphoid-like PC represent the majority of PC and include pTalpha-, CD3epsilon-, and CD7-expressing cells. They exhibit TCR-beta gene loci in germline configuration and show low allostimulatory capacity, but produce type I IFN upon virus infection and can be differentiated in vitro into potent APC. Myeloid-like PC represent a minor fraction of the total PC population. They exhibit a striking PC/myeloid DC intermediate phenotype (CD5(+)CD11c(low)CD45RA(low)CD45RO(-)CD101(+)), produce proinflammatory cytokines, and do not require in vitro maturation to act as potent APCs. We propose that, rather than forming a lineage, PC might represent a population of lymphoid cells undergoing an in vivo cell fate conversion from a lymphoid to a myeloid cell type.
Collapse
MESH Headings
- Antigen-Presenting Cells/cytology
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Antigens, CD1/biosynthesis
- Blood Cell Count
- CD3 Complex/biosynthesis
- CD40 Ligand/pharmacology
- CD5 Antigens/biosynthesis
- CD56 Antigen/biosynthesis
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cell Lineage/genetics
- Cell Lineage/immunology
- Cell Separation
- Cells, Cultured
- Dendritic Cells/cytology
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/virology
- Gene Rearrangement, beta-Chain T-Cell Antigen Receptor
- Genetic Markers
- Glycoproteins/biosynthesis
- HLA-DR Antigens/biosynthesis
- Hematopoietic Stem Cells/cytology
- Hematopoietic Stem Cells/immunology
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/virology
- Herpesvirus 1, Human/immunology
- Humans
- Immunophenotyping
- Injections, Subcutaneous
- Interferon Type I/biosynthesis
- Interleukin-1/biosynthesis
- Interleukin-3/pharmacology
- Interleukin-3 Receptor alpha Subunit
- Interleukin-6/biosynthesis
- Ligands
- Membrane Glycoproteins/biosynthesis
- Membrane Proteins/administration & dosage
- Myeloid Cells/cytology
- Myeloid Cells/immunology
- Myeloid Cells/metabolism
- Plasma Cells/cytology
- Plasma Cells/immunology
- Plasma Cells/metabolism
- Plasma Cells/virology
- Receptors, Antigen, T-Cell/biosynthesis
- Receptors, Antigen, T-Cell, alpha-beta/biosynthesis
- Receptors, Antigen, T-Cell, gamma-delta/biosynthesis
- Receptors, Interleukin-3/biosynthesis
Collapse
|
17
|
Petrella T, Comeau MR, Maynadié M, Couillault G, De Muret A, Maliszewski CR, Dalac S, Durlach A, Galibert L. 'Agranular CD4+ CD56+ hematodermic neoplasm' (blastic NK-cell lymphoma) originates from a population of CD56+ precursor cells related to plasmacytoid monocytes. Am J Surg Pathol 2002; 26:852-62. [PMID: 12131152 DOI: 10.1097/00000478-200207000-00003] [Citation(s) in RCA: 183] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In 1999, we reported seven cases of an unusual hematologic malignancy with primary cutaneous presentation that appeared as a distinct clinicopathologic entity characterized by medium-sized tumor cells with a peculiar CD3- CD4+ CD56+ CD43+ HLA-DR+ cell surface phenotype. Because the origin of tumor cells was not clear and they exhibited a nonlineage-specific phenotype, we hypothesized that such tumors likely originated from hematologic-myeloid precursor cells and were tentatively assigned the designation "agranular CD4+ CD56+ hematodermic neoplasms." In the present study we report 14 cases (seven already reported and seven additional cases) of these tumors, and simultaneously we present now a rare population of cells that we have identified in the peripheral blood of healthy volunteers treated with Flt3 ligand. These cells express all the characteristic markers of CD4+ CD56+ hematodermic neoplasms. This population appears to be related to plasmacytoid monocytes because they also expressed CD68 and bright levels of CD123. To confirm the relationship between these normal cells and CD4+ CD56+ hematodermic neoplasms, we conducted an extensive comparative phenotypic study. Results show that these two cell types are indeed related because they share many phenotypic features, including the presence of CD4, CD56, CD43, CD68, and HLA-DR and the absence of other T, B, NK, or myelomonocytic markers. More importantly, we found that the bright expression of CD123 by immunohistochemistry is a distinctive characteristic of CD4+ CD56+ hematodermic neoplasms because all (n = 14) cases expressed this marker, whereas only two specimens in a control panel comprising 30 samples of related tumors expressed comparable levels of CD123. We therefore propose that oncogenic transformation of NCAM-expressing plasmacytoid monocyte-like cells may lead to "agranular CD4+ CD56+ hematodermic neoplasm."
Collapse
Affiliation(s)
- Tony Petrella
- Centre de Pathologie of Dijon and the Department of Pathology, Dijon University Hospital, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Gary-Gouy H, Lebon P, Dalloul AH. Type I interferon production by plasmacytoid dendritic cells and monocytes is triggered by viruses, but the level of production is controlled by distinct cytokines. J Interferon Cytokine Res 2002; 22:653-9. [PMID: 12162875 DOI: 10.1089/10799900260100132] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
The principal interferon-alpha/beta (IFN-I)-producing cells are plasmacytoid dendritic cell (PDC) precursors belonging to the lymphoid lineage. Monocytes that can differentiate into dendritic cells (DC) also produce IFN-I, although much less than PDC, after interaction with infectious agents. We show that whereas viruses trigger these cells to produce IFN-I, the amount of IFN is tightly controlled by cytokines. Monocytes produced IFN-I in response to Sendai virus (SV) infection, and PDC responded to both SV and herpes simplex virus (HSV). All cytokines tested failed to induce production of IFN-I in the absence of infection. However, among 18 relevant cytokines, incubation of PDC with interleukin-4 (IL-4), IL-15, and IL-7 alone or in combination with IL-3 before infection, enhanced IFN-I secretion. At variance, IL-12 alone or in synergy with granulocyte-macrophage colony-stimulating factor (GM-CSF) was active on SV-infected but not on HSV-infected monocytes. Tumor necrosis factor-alpha (TNF-alpha) and IL-4 inhibited IFN-I production by PDC and monocytes, respectively, and IL-10 strongly inhibited IFN-I production in both cell lineages. The response of PDC to IL-7 and IL-15, which also activate natural killer (NK) cell maturation, further emphasizes the cooperation between these two cell subsets in the control of innate immunity.
Collapse
Affiliation(s)
- Hélène Gary-Gouy
- Laboratoire d'Immunologie, INSERM U 543, Hôpital Pitié-Salpêtrière, Université de Paris VI, 75013 Paris, France
| | | | | |
Collapse
|
19
|
Gounari F, Aifantis I, Martin C, Fehling HJ, Hoeflinger S, Leder P, von Boehmer H, Reizis B. Tracing lymphopoiesis with the aid of a pTalpha-controlled reporter gene. Nat Immunol 2002; 3:489-96. [PMID: 11927910 DOI: 10.1038/ni778] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A transgenic reporter mouse strain, which expressed the human CD25 (hCD25) surface marker as a reporter under the control of the pre-T cell receptor alpha(pTalpha) promoter, was used to identify lymphoid precursors that expressed pTalpha intracellularly. The hCD25 reporter marked intra- and extrathymic precursors of lymphocytes but not myeloid cells. The earliest intrathymic precursors were CD4(lo)CD8(-)CD25(-)CD44(+)c-Kit(+) cells that expressed elevated levels of Notch-1 mRNA. Clonogenic assays showed that the extrathymic precursors were common lymphoid progenitors (CLPs) that included CD19(-), B220(+), Thy1(+) and CD4(+) cells. Thus, the pTalpha reporter can be used to trace lymphopoiesis between CLPs and alphabeta T cells. The slower extinction of the hCD25 reporter compared to pTalpha enabled us to define points at which pTalpha(-) lineages branched off.
Collapse
MESH Headings
- Animals
- Flow Cytometry
- Gene Expression Regulation
- Hematopoiesis/immunology
- Humans
- Lymphocytes/cytology
- Lymphocytes/immunology
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Membrane Proteins/analysis
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Phenotype
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Receptor, Notch1
- Receptors, Antigen, T-Cell, alpha-beta
- Receptors, Cell Surface
- Receptors, Interleukin-2/immunology
- Reverse Transcriptase Polymerase Chain Reaction
- Stem Cells
- Transcription Factors
Collapse
Affiliation(s)
- Fotini Gounari
- Department of Pathology, Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Dendritic cells (DCs) are bone marrow-derived cells of both lymphoid and myeloid stem cell origin that populate all lymphoid organs including the thymus, spleen, and lymph nodes, as well as nearly all nonlymphoid tissues and organs. Although DCs are a moderately diverse set of cells, they all have potent antigen-presenting capacity for stimulating naive, memory, and effector T cells. DCs are members of the innate immune system in that they can respond to dangers in the host environment by immediately generating protective cytokines. Most important, immature DCs respond to danger signals in the microenvironment by maturing, i.e., differentiating, and acquiring the capacity to direct the development of primary immune responses appropriate to the type of danger perceived. The powerful adjuvant activity that DCs possess in stimulating specific CD4 and CD8 T cell responses has made them targets in vaccine development strategies for the prevention and treatment of infections, allograft reactions, allergic and autoimmune diseases, and cancer. This review addresses the origins and migration of DCs to their sites of activity, their basic biology as antigen-presenting cells, their roles in important human diseases and, finally, selected strategies being pursued to harness their potent antigen-stimulating activity.
Collapse
Affiliation(s)
- Mary F Lipscomb
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, New Mexico 87131-5301, USA.
| | | |
Collapse
|
21
|
Mohty M, Jarrossay D, Lafage-Pochitaloff M, Zandotti C, Brière F, de Lamballeri XN, Isnardon D, Sainty D, Olive D, Gaugler B. Circulating blood dendritic cells from myeloid leukemia patients display quantitative and cytogenetic abnormalities as well as functional impairment. Blood 2001; 98:3750-6. [PMID: 11739182 DOI: 10.1182/blood.v98.13.3750] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Dendritic cells (DCs) are responsible for the initiation of immune responses. Two distinct subsets of blood DCs have been characterized thus far. Myeloid DCs (MDCs) and plasmacytoid monocytes (PDCs) were shown to be able to promote polarization of naive T cells. This study shows a dramatic quantitative imbalance in both circulating blood DC subsets in 37 patients with acute myeloid leukemias. Eleven patients (30%) displayed a normal quantitative profile (MDC mean, 0.37% +/- 0.21%; range, 0.01% to 0.78%; PDC mean, 0.21% +/- 0.24%; range, 0.04% to 0.62%), whereas 22 (59%) showed a tremendous expansion of MDCs (9 patients: mean, 16.76% +/- 14.03%; range, 1.36% to 41%), PDCs (4 patients: mean, 7.28% +/- 6.84%; range, 1% to 14%), or both subsets (9 patients: MDC mean, 10.86% +/- 12.36%; range, 1.02% to 37.1%; PDC mean, 4.25% +/- 3.78%; range, 1.14% to 13.04%). Finally, in 4 patients (11%), no DC subsets were detectable. Both MDC and PDC subsets exhibited the original leukemic chromosomal abnormality. Ex vivo, leukemic PDCs, but not leukemic MDCs, had impaired capacity for maturation and decreased allostimulatory activity. Also, leukemic PDCs were altered in their ability to secrete interferon-alpha. These data provide evidence that DC subsets in vivo may be affected by leukemogenesis and may contribute to leukemia escape from immune control.
Collapse
Affiliation(s)
- M Mohty
- Laboratoire d'Immunologie des Tumeurs, Université de la Méditerranée, Marseille, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
The generation of a peripheral T-cell pool is essential for normal immune system function. CD4+ and CD8+ T cells are produced most efficiently in the thymus, which provides a complexity of discrete cellular microenvironments. Specialized stromal cells, that make up such microenvironments, influence each stage in the maturation programme of immature T-cell precursors. Progress has recently been made in elucidating events that regulate the development of intrathymic microenvironments, as well as mechanisms of thymocyte differentiation. It is becoming increasingly clear that the generation and maintenance of thymic environments that are capable of supporting efficient T-cell development, requires complex interplay between lymphoid and stromal compartments of the thymus.
Collapse
Affiliation(s)
- G Anderson
- Department of Anatomy, MRC Centre for Immune Regulation, University of Birmingham, Edgbaston, UK.
| | | |
Collapse
|
23
|
Abstract
Plasmacytoid monocytes/T cells were first described in 1958, yet their origin and function have remained enigmatic. Recently a series of publications brought these cells to the forefront of immunological research. Indeed, plasmacytoid monocytes/T-cells contain natural type-I interferon producing cells and can differentiate in vitro into dendritic cells (DC). It has been proposed that plasmacytoid monocytes/T-cells represent a distinct lineage of cells whose fate it is to differentiate into dendritic cells. Herein we will review recent advances in our understanding of plasmacytoid monocytes/T cells and highlight arguments in favor or against this lineage hypothesis. We propose that plasmacytoid monocytes/T cells represent a composite group of both myeloid and lymphoid early-committed cells that are characterized by their ability to differentiate in vitro into DC.
Collapse
Affiliation(s)
- L Galibert
- Discovery Research, Immunex Corporation, 51 University Street, Seattle, WA 98101-2936, USA.
| | | | | |
Collapse
|
24
|
Bank I, Amariglio N, Reshef A, Hardan I, Confino Y, Trau H, Shtrasburg S, Langevitz P, Monselise Y, Shalit M, Rechavi G. The hypereosinophilic syndrome associated with CD4+CD3- helper type 2 (Th2) lymphocytes. Leuk Lymphoma 2001; 42:123-33. [PMID: 11699199 DOI: 10.3109/10428190109097684] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We describe herein the clinical and laboratory manifestations of a unique group of patients (pts) presenting with hypereosinophilic syndrome (HES) who were treated in our medical centers for 4-13 years. Skin biopsies, flow cytometry of peripheral blood mononuclear cells (PBMC), assays for cytokines and immunoglobulin (Ig) production in vitro, and Southern blots of T-cell receptor (TCR) genes were performed. All four pts had a persistent hypereosinophilia (> 1.9 x 10(9)/L) and chronic skin rash. Three of four had elevated IgE, thrombotic manifestations and lung involvement (asthma and/or infiltrates), and one had deforming sero-negative arthritis of the hands. 66-95% of their peripheral T-cells expressed CD4 but not CD3 or TCR molecules on the cell surface membrane. Activated CD4+CD3- cells secreted interleukin (IL)-4 and/or 5, and were required for maximal IgE secretion by autologous B-cells. Two pts had evidence of rearrangement of TCR genes of the CD4+CD3- cells, one of whom died of anaplastic lymphoma. In conclusion, HES with CD4+CD3- lymphocytosis may be associated with high serum IgE, dermatological, pulmonary, thrombotic and rheumatic manifestations which may be due to Th2 effects of CD4+CD3- cells migrating to end organs. Fatal systemic lymphoid malignancy may also develop in some pts with monoclonal expansion of the CD4+CD3- T-cells.
Collapse
Affiliation(s)
- I Bank
- Department of Medicine, Laboratory for Immunoregulation, Chaim Sheba Medical Center, Tel-Hashomer 52621 Israel.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Hochrein H, Shortman K, Vremec D, Scott B, Hertzog P, O'Keeffe M. Differential production of IL-12, IFN-alpha, and IFN-gamma by mouse dendritic cell subsets. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:5448-55. [PMID: 11313382 DOI: 10.4049/jimmunol.166.9.5448] [Citation(s) in RCA: 376] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Dendritic cells (DC) not only stimulate T cells effectively but are also producers of cytokines that have important immune regulatory functions. In this study we have extended information on the functional differences between DC subpopulations to include differences in the production of the major immune-directing cytokines IL-12, IFN-alpha, and IFN-gamma. Splenic CD4(-)8(+) DC were identified as the major IL-12 producers in response to microbiological or T cell stimuli when compared with splenic CD4(-)8(-) or CD4(+)8(-) DC; however, all three subsets of DC showed similar IL-12 regulation and responded with increased IL-12 p70 production if IL-4 was present during stimulation. High level CD8 expression also correlated with extent of IL-12 production for DC isolated from thymus and lymph nodes. By using gene knockout mice we ruled out any role for CD8alpha itself, or of priming by T cells, on the superior IL-12-producing capacity of the CD8(+) DC. Additionally, CD8(+) DC were identified as the major producers of IFN-alpha compared with the two CD8(-) DC subsets, a finding that suggests similarity to the human plasmacytoid DC lineage. In contrast, the CD4(-)8(-) DC produced much more IFN-gamma than the CD4(-)8(+) or the CD4(+)8(-) DC under all conditions tested.
Collapse
Affiliation(s)
- H Hochrein
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.
| | | | | | | | | | | |
Collapse
|
26
|
Bendriss-Vermare N, Barthélémy C, Durand I, Bruand C, Dezutter-Dambuyant C, Moulian N, Berrih-Aknin S, Caux C, Trinchieri G, Brière F. Human thymus contains IFN-alpha-producing CD11c(-), myeloid CD11c(+), and mature interdigitating dendritic cells. J Clin Invest 2001; 107:835-44. [PMID: 11285302 PMCID: PMC199577 DOI: 10.1172/jci11734] [Citation(s) in RCA: 152] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Three distinct dendritic cell (DC) subsets capable of stimulating allogeneic naive T cells were isolated from human thymus. The most abundant subset was represented by plasmacytoid DCs (pDCs), which secreted high amounts of IFN-alpha upon stimulation with inactivated influenza virus and thus likely correspond to the recently identified peripheral blood natural IFN-alpha/beta-producing cells (IPCs). Like those latter cells, thymic pDCs had distinctive phenotypic features (i.e., Lin(-), HLA-DR(int), IL-3R alpha(hi), CD45RA(hi), CD11c(-), CD13(-), and CD33(lo)) and developed into mature DCs upon culture in IL-3 and CD40L. Of the two other DC subsets, one displayed a phenotype of immature myeloid DCs (imDCs) (HLA-DR(int), CD11c(+), CD13(+), CD33(+)), and the other represented HLA-DR(hi) CD11c(+) mature DCs (mDCs). Since they also expressed DC-LAMP, these mDCs appear to correspond to interdigitating dendritic cells (IDCs). Thymic pDCs, but not myeloid imDCs, strongly expressed lymphoid-specific transcripts such as pre-T alpha, lambda-like, and Spi-B, thereby suggesting a possible lymphoid origin. The detection of Spi-B mRNA, not only upon in vitro maturation of pDCs, but also in freshly purified IDCs, suggests that in vivo pDCs may differentiate into IDCs.
Collapse
Affiliation(s)
- N Bendriss-Vermare
- Schering-Plough, Laboratory for Immunological Research, Dardilly, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Toor AA, Lund TC, Miller JS. T-cell factor-1 expression during human natural killer cell development and in circulating CD56(+) bright natural killer cells. Exp Hematol 2001; 29:499-506. [PMID: 11301190 DOI: 10.1016/s0301-472x(00)00680-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Transcription factors are essential to govern differentiation along the lymphoid lineage from uncommitted hematopoietic stem cells. Although many of these transcription factors have putative roles based on murine knockout experiments, their function in human lymphoid development is less known and was studied further. Transcription factor expression in fresh and cultured adult human bone marrow and umbilical cord blood progenitors was evaluated. We found that fresh CD34(+)Lin(-) cells that are human leukocyte antigen (HLA)-DR(-) or CD38(-) constitutively express GATA-3 but not T-cell factor-1 (TCF-1) or Id-3. Culture with the murine fetal liver cell line AFT024 and defined cytokines was capable of inducing TCF-1 mRNA. However, no T-cell receptor gene rearrangement was identified in cultured progeny. Id-3, a basic helix loop helix factor with dominant negative function for T-cell differentiation transcription factors, also was upregulated and may explain unsuccessful T-cell maturation. To better understand the developmental link between natural killer (NK) cells derived from progenitors, we studied NK cell subsets circulating in blood. CD56(+bright), but not CD56(+dim), NK cells constitutively express TCF-1 by reverse transcriptase polymerase chain reaction and Western blot analysis. The TCF-1 isoform found in CD56(+bright) cells, which express lectin but not immunoglobulin class I recognizing inhibitory receptors, was identical to that induced in NK cell differentiation culture and was distinctly different from isoforms in T cells. These results suggest that TCF-1 does not target human killer immunoglobulin receptor genes, TCF-1 is uniquely expressed in circulating CD56(+bright) NK cells, and specific TCF-1 isoforms may play an important role in regulating NK differentiation from a common NK/T-cell progenitor.
Collapse
Affiliation(s)
- A A Toor
- Department of Medicine, University of Minnesota Cancer Center, Harvard Street at East River Road, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
28
|
Vandenabeele S, Hochrein H, Mavaddat N, Winkel K, Shortman K. Human thymus contains 2 distinct dendritic cell populations. Blood 2001; 97:1733-41. [PMID: 11238115 DOI: 10.1182/blood.v97.6.1733] [Citation(s) in RCA: 111] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In this study, 2 distinct populations of mature dendritic cells (DCs) were identified in the human thymus. The major population is CD11b-, CD11c+, and CD45RO(low) and does not express myeloid-related markers. It displays all the characteristics of mature DCs with a typical dendritic morphology, high surface levels of HLA-DR, CD40, CD83, and CD86, and expression of DC-lysosome-associated membrane glycoprotein messenger RNA (mRNA). In addition, CD11b- thymic DCs do not express macrophage inflammatory protein-1alpha (MIP-1alpha) mRNA, but express thymus-expressed chemokine (TECK) mRNA and are able to secrete bioactive interleukin 12 (IL-12) upon stimulation. In contrast, the minor and variable thymic DC population is CD11b+, CD11c(high), and CD45RO(high) and comprises CD83+CD14- mature and CD83- CD14+ immature DCs. It expresses macrophage-colony stimulating factor receptor, MIP-1alpha mRNA and high amounts of decysin mRNA after CD40 activation, but does not express TECK and is a weak bioactive IL-12 producer. Also identified were the IL-3Ralpha(high) plasmacytoid cells, which are present in the thymic cortex and medulla. Upon culture with IL-3, granulocyte/macrophage-colony stimulating factor, and CD40 ligand, the plasmacytoid cells can adopt a phenotype resembling that of freshly isolated CD11b- thymic DCs. However, these plasmacytoid-derived DCs fail to secrete bioactive IL-12; therefore, conclusions cannot be made about a direct relation between thymic plasmacytoid cells and CD11b- DCs. Whereas CD11b+ thymic DCs appear to be related to tonsillar germinal-center DCs, the major CD11b- IL-12-secreting human thymus DC population has similarities to mouse CD11b- CD8+ DCs.
Collapse
Affiliation(s)
- S Vandenabeele
- The Walter and Eliza Hall Institute of Medical Research, P.O. Royal Melbourne Hospital, 3050 Melbourne, Victoria, Australia.
| | | | | | | | | |
Collapse
|
29
|
Grage‐Griebenow E, Flad H, Ernst M. Heterogeneity of human peripheral blood monocyte subsets. J Leukoc Biol 2001. [DOI: 10.1189/jlb.69.1.11] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
| | - H.‐D. Flad
- Immunology and Cell Biology, Research Center Borstel, Germany
| | - M. Ernst
- Immunology and Cell Biology, Research Center Borstel, Germany
| |
Collapse
|
30
|
Pulendran B, Banchereau J, Maraskovsky E, Maliszewski C. Modulating the immune response with dendritic cells and their growth factors. Trends Immunol 2001; 22:41-7. [PMID: 11286691 DOI: 10.1016/s1471-4906(00)01794-4] [Citation(s) in RCA: 194] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Different subsets of dendritic cells (DCs) appear to play a role in determining the specific cytokines secreted by T helper (Th) cells. A model is proposed that links together factors such as the pathogen, microenvironment, DCs and T cells in a mechanism that results in a flexible determination of T-cell polarization.
Collapse
Affiliation(s)
- B Pulendran
- Baylor Institute for Immunology, Dallas, TX 75204, USA.
| | | | | | | |
Collapse
|
31
|
Blom B, Ho S, Antonenko S, Liu YJ. Generation of interferon alpha-producing predendritic cell (Pre-DC)2 from human CD34(+) hematopoietic stem cells. J Exp Med 2000; 192:1785-96. [PMID: 11120775 PMCID: PMC2213503 DOI: 10.1084/jem.192.12.1785] [Citation(s) in RCA: 175] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Upon viral stimulation, the natural interferon (IFN)-alpha/beta-producing cells (IPCs; also known as pre-dendritic cells (DCs 2) in human blood and peripheral lymphoid tissues rapidly produce huge amounts of IFN-alpha/beta. After performing this innate antiviral immune response, IPCs can differentiate into DCs and strongly stimulate T cell-mediated adaptive immune responses. Using four-color immunofluorescence flow cytometry, we have mapped the developmental pathway of pre-DC2/IPCs from CD34(+) hematopoietic stem cells in human fetal liver, bone marrow, and cord blood. At least four developmental stages were identified, including CD34(++)CD45RA(-) early progenitor cells, CD34(++)CD45RA(+) late progenitor cells, CD34(+)CD45RA(++)CD4(+)interleukin (IL)-3Ralpha(++) pro-DC2, and CD34(-)CD45RA(++) CD4(+)IL-3Ralpha(++) pre-DC2/IPCs. Pro-DC2s have already acquired the capacity to produce large amounts of IFN-alpha/beta upon viral stimulation and to differentiate into DCs in culture with IL-3 and CD40 ligand. CD34(++)CD45RA(-) early progenitor cells did not have the capacity to produce large amounts of IFN-alpha/beta in response to viral stimulation; however, they can be induced to undergo proliferation and differentiation into IPCs/pre-DC2 in culture with FLT3 ligand.
Collapse
Affiliation(s)
- Bianca Blom
- Department of Immunobiology, DNAX Research Institute of Molecular and Cellular Biology, Palo Alto, California 94304
| | - Stephen Ho
- Department of Immunobiology, DNAX Research Institute of Molecular and Cellular Biology, Palo Alto, California 94304
| | - Svetlana Antonenko
- Department of Immunobiology, DNAX Research Institute of Molecular and Cellular Biology, Palo Alto, California 94304
| | - Yong-Jun Liu
- Department of Immunobiology, DNAX Research Institute of Molecular and Cellular Biology, Palo Alto, California 94304
| |
Collapse
|
32
|
Dzionek A, Fuchs A, Schmidt P, Cremer S, Zysk M, Miltenyi S, Buck DW, Schmitz J. BDCA-2, BDCA-3, and BDCA-4: three markers for distinct subsets of dendritic cells in human peripheral blood. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:6037-46. [PMID: 11086035 DOI: 10.4049/jimmunol.165.11.6037] [Citation(s) in RCA: 951] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We have generated a panel of mAbs that identify three presumably novel human dendritic cell Ags: BDCA-2, BDCA-3, and BDCA-4. In blood, BDCA-2 and BDCA-4 are expressed on CD11c(-) CD123(bright) plasmacytoid dendritic cells, whereas BDCA-3 is expressed on small population of CD11c(+) CD123(-) dendritic cells. All three Ags are not detectable on a third blood dendritic cell population, which is CD1c(+) CD11c(bright) CD123(dim), or on any other cells in blood. BDCA-4 is also expressed on monocyte-derived and CD34(+) cell-derived dendritic cells. Expression of all three Ags dramatically changes once blood dendritic cells undergo in vitro maturation. BDCA-2 is completely down-regulated on plasmacytoid CD11c(-) CD123(bright) dendritic cells, expression of BDCA-3 is up-regulated on both plasmacytoid CD11c(-) CD123(bright) dendritic cells and CD1c(+) CD11c(bright) CD123(dim) dendritic cells, and expression of BDCA-4 is up-regulated on CD1c(+) CD11c(bright) CD123(dim) dendritic cells. BDCA-2 is rapidly internalized at 37 degrees C after mAb labeling. The three presumably novel Ags serve as specific markers for the respective subpopulations of blood dendritic cells in fresh blood and will be of great value for their further analysis and to evaluate their therapeutic potential.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/metabolism
- Antibody Specificity
- Antigen-Antibody Reactions
- Antigens, CD
- Antigens, CD1/biosynthesis
- Antigens, CD34/biosynthesis
- Antigens, Differentiation/biosynthesis
- Antigens, Differentiation/blood
- Antigens, Differentiation/immunology
- Antigens, Differentiation/metabolism
- Antigens, Surface/biosynthesis
- Biomarkers/blood
- Cell Separation
- Cells, Cultured
- Dendritic Cells/cytology
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Endocytosis/immunology
- Female
- Flow Cytometry
- Histocompatibility Antigens Class II/biosynthesis
- Humans
- Immunoglobulins/biosynthesis
- Immunophenotyping
- Lymphocytes/cytology
- Lymphocytes/immunology
- Lymphocytes/metabolism
- Membrane Glycoproteins/biosynthesis
- Mice
- Mice, Inbred BALB C
- Monocytes/immunology
- Monocytes/metabolism
- Plasma Cells/cytology
- Plasma Cells/immunology
- Plasma Cells/metabolism
- Staining and Labeling
- CD83 Antigen
Collapse
Affiliation(s)
- A Dzionek
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany. AmCell Corporation, Sunnyvale, CA 94089, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Schmitt C, Fohrer H, Beaudet S, Palmer P, Alpha M, Canque B, Gluckman JC, Dalloul AH. Identification of mature and immature human thymic dendritic cells that differentially express HLA‐DR and interleukin‐3 receptor
in vivo. J Leukoc Biol 2000. [DOI: 10.1189/jlb.68.6.836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Christian Schmitt
- UMR CNRS 7627, Hopital Pitié‐Salpêtrière, Paris, France
- Laboratoire de Virologie, Faculté de Médecine Cochin‐Paris V, Paris, France
- ESA 7087 UP6‐CNRS and Laboratoire d’Immunologie et Immunopathologie de l’ Ecole Pratique des Hautes Etudes, Paris, France
| | - Hélène Fohrer
- UMR CNRS 7627, Hopital Pitié‐Salpêtrière, Paris, France
- Laboratoire de Virologie, Faculté de Médecine Cochin‐Paris V, Paris, France
- ESA 7087 UP6‐CNRS and Laboratoire d’Immunologie et Immunopathologie de l’ Ecole Pratique des Hautes Etudes, Paris, France
| | - Sylvie Beaudet
- UMR CNRS 7627, Hopital Pitié‐Salpêtrière, Paris, France
- Laboratoire de Virologie, Faculté de Médecine Cochin‐Paris V, Paris, France
- ESA 7087 UP6‐CNRS and Laboratoire d’Immunologie et Immunopathologie de l’ Ecole Pratique des Hautes Etudes, Paris, France
| | - Pierre Palmer
- Laboratoire de Virologie, Faculté de Médecine Cochin‐Paris V, Paris, France
| | - Marie‐José Alpha
- UMR CNRS 7627, Hopital Pitié‐Salpêtrière, Paris, France
- Laboratoire de Virologie, Faculté de Médecine Cochin‐Paris V, Paris, France
- ESA 7087 UP6‐CNRS and Laboratoire d’Immunologie et Immunopathologie de l’ Ecole Pratique des Hautes Etudes, Paris, France
| | - Bruno Canque
- ESA 7087 UP6‐CNRS and Laboratoire d’Immunologie et Immunopathologie de l’ Ecole Pratique des Hautes Etudes, Paris, France
| | - Jean Claude Gluckman
- ESA 7087 UP6‐CNRS and Laboratoire d’Immunologie et Immunopathologie de l’ Ecole Pratique des Hautes Etudes, Paris, France
| | - Ali H. Dalloul
- UMR CNRS 7627, Hopital Pitié‐Salpêtrière, Paris, France
- Laboratoire de Virologie, Faculté de Médecine Cochin‐Paris V, Paris, France
- ESA 7087 UP6‐CNRS and Laboratoire d’Immunologie et Immunopathologie de l’ Ecole Pratique des Hautes Etudes, Paris, France
| |
Collapse
|
34
|
Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu YJ, Pulendran B, Palucka K. Immunobiology of dendritic cells. Annu Rev Immunol 2000; 18:767-811. [PMID: 10837075 DOI: 10.1146/annurev.immunol.18.1.767] [Citation(s) in RCA: 4768] [Impact Index Per Article: 198.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DCs) are antigen-presenting cells with a unique ability to induce primary immune responses. DCs capture and transfer information from the outside world to the cells of the adaptive immune system. DCs are not only critical for the induction of primary immune responses, but may also be important for the induction of immunological tolerance, as well as for the regulation of the type of T cell-mediated immune response. Although our understanding of DC biology is still in its infancy, we are now beginning to use DC-based immunotherapy protocols to elicit immunity against cancer and infectious diseases.
Collapse
Affiliation(s)
- J Banchereau
- Baylor Institute for Immunology Research, Dallas, Texas 75204, USA.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Abstract
Peripheral blood stem cells (PBSC) obtained from granulocyte-colony stimulating factor (G-CSF)-mobilized donors are increasingly used for allogeneic transplantation. Despite a 10-fold higher dose of transplanted T cells, acute graft-versus-host disease (GVHD) does not develop in higher proportion in recipients of PBSC than in recipients of marrow. T cells from G-CSF-treated experimental animals preferentially produce IL-4 and IL-10, cytokines characteristic of Th2 responses, which are associated with diminished GVHD-inducing ability. We hypothesized that G-CSF-mobilized PBSC contain antigen-presenting cells, which prime T-lymphocytes to produce Th2 cytokines. Two distinct lineages of dendritic cells (DC) have been described in humans, DC1 and DC2, according to their ability to induce naive T-cell differentiation to Th1 and Th2 effector cells, respectively. We have used multicolor microfluorometry to enumerate DC1 and DC2 in the peripheral blood of normal donors. G-CSF treatment with 10 to 16 μg/kg per day for 5 days increased peripheral blood DC2 counts from a median of 4.9 × 106/L to 24.8 × 106/L (P = .0009), whereas DC1 counts did not change. Purified DC1, from either untreated or G-CSF treated donors, induced the proliferation of allogeneic naive T cells, but fresh DC2 were poor stimulators. Tumor necrosis factor- (TNF-)-activated DC1 induced allogeneic naive T cells to produce IFN-γ, which is typical of Th1 responses, whereas TNF--activated DC2 induced allogeneic naive T cells to produce IL-4 and IL-10, which are typical of Th2 responses. PBSC transplants contained higher doses of DC2 than marrow transplants (median, 2.4 × 106/kg versus 0.5 × 106/kg) (P = .006), whereas the dose of DC1 was comparable. Thus, it is conceivable that transplantation of G-CSF-stimulated PBSC does not result in overwhelming acute GVHD because the graft contains predominantly Th2-inducing DC. Adoptive transfer of purified DC2 may be exploited to induce immune deviation after transplantation of hematopoietic stem cells or organ allografts.
Collapse
|
36
|
Abstract
Peripheral blood stem cells (PBSC) obtained from granulocyte-colony stimulating factor (G-CSF)-mobilized donors are increasingly used for allogeneic transplantation. Despite a 10-fold higher dose of transplanted T cells, acute graft-versus-host disease (GVHD) does not develop in higher proportion in recipients of PBSC than in recipients of marrow. T cells from G-CSF-treated experimental animals preferentially produce IL-4 and IL-10, cytokines characteristic of Th2 responses, which are associated with diminished GVHD-inducing ability. We hypothesized that G-CSF-mobilized PBSC contain antigen-presenting cells, which prime T-lymphocytes to produce Th2 cytokines. Two distinct lineages of dendritic cells (DC) have been described in humans, DC1 and DC2, according to their ability to induce naive T-cell differentiation to Th1 and Th2 effector cells, respectively. We have used multicolor microfluorometry to enumerate DC1 and DC2 in the peripheral blood of normal donors. G-CSF treatment with 10 to 16 μg/kg per day for 5 days increased peripheral blood DC2 counts from a median of 4.9 × 106/L to 24.8 × 106/L (P = .0009), whereas DC1 counts did not change. Purified DC1, from either untreated or G-CSF treated donors, induced the proliferation of allogeneic naive T cells, but fresh DC2 were poor stimulators. Tumor necrosis factor- (TNF-)-activated DC1 induced allogeneic naive T cells to produce IFN-γ, which is typical of Th1 responses, whereas TNF--activated DC2 induced allogeneic naive T cells to produce IL-4 and IL-10, which are typical of Th2 responses. PBSC transplants contained higher doses of DC2 than marrow transplants (median, 2.4 × 106/kg versus 0.5 × 106/kg) (P = .006), whereas the dose of DC1 was comparable. Thus, it is conceivable that transplantation of G-CSF-stimulated PBSC does not result in overwhelming acute GVHD because the graft contains predominantly Th2-inducing DC. Adoptive transfer of purified DC2 may be exploited to induce immune deviation after transplantation of hematopoietic stem cells or organ allografts.
Collapse
|
37
|
Abstract
We have characterized dendritic cell precursors (pre-DC) in the human thymus. These CD1a−CD3−CD4+CD8−cells express high levels of interleukin-3R (IL-3R) on the membrane and are able to develop into mature DC upon culture with IL-3 and CD40 ligation. The DC precursors are predominantly located in the thymic medulla. Interestingly, the pre-DC express pT mRNA, which is also present in CD1a+CD3−CD4+ CD8−pre-T cells. Yet, the pre-DC lack expression of recombination activating gene-1 mRNA and fail to develop into T cells in appropriate assays. The thymic pre-DC are very similar to the recently characterized pre-DC found in the T cell areas of the tonsil, and it is suggested that these pre-DC populations are of lymphoid origin.
Collapse
|
38
|
Abstract
In the present review, a series of studies on the origins of dendritic cells of mice and humans are summarized. Several subsets of mature dendritic cells found in vivo are described and these may correspond to distinct lineages. There is evidence that some dendritic cells are myeloid-derived and that others are lymphoid-derived. The different ways of generating dendritic cells are examined and an attempt to reconcile the differences seen using mouse and human culture models is made. The particular case of Langerhans cells is discussed and an historical overview of the biology of the plasmacytoid T cells, which may represent a distinct 'lymphoid-related' dendritic cell lineage, is given. It is concluded that three or four different pathways lead to the development of different subtypes of dendritic cells.
Collapse
Affiliation(s)
- S Vandenabeele
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.
| | | |
Collapse
|
39
|
|
40
|
Differentiation in Culture of Murine Primitive Lymphohematopoietic Progenitors Toward T-Cell Lineage. Blood 1999. [DOI: 10.1182/blood.v93.12.4187] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Abstract
Earlier, we described a stromal cell-free two-step clonal culture system in which murine primitive lymphohematopoietic progenitors produce myeloid and B-lymphoid lineage cells. In the same culture T-cell potential of the progenitors was maintained. We now report that, in addition to myeloid and B-lymphoid cells, putative T-cell progenitors are also produced in culture. Lineage-negative (Lin−) Ly-6A/E+ c-kit+ bone marrow cells from 5-fluorouracil–treated mice were cultured in methylcellulose in the presence of SF (Steel factor), interleukin (IL)-11, and IL-7, and the resulting primary colonies were picked and pooled. When injected into severe combined immune deficiency (scid) mice, the pooled cells reconstituted the T-cell compartment of the scid mice earlier than freshly prepared primitive marrow cells. This reconstitution activity of the pooled primary colony cells was enriched in the Ly-6A/E+ and FcγRII/III−/low cell fractions. Reverse transcriptase-polymerase chain reaction (RT-PCR) and DNA-PCR analyses showed that some of the primary colony cells are differentiated sufficiently to express messenger RNA (mRNA) of T-cell receptor (TCR) β-chain and pre-TCR alpha (pT) and, although not frequently, to perform Dβ-Jβ rearrangement of the TCR gene. Micromanipulation studies confirmed the clonal origin of myeloid lineage cells and the cells positive for the T-cell–specific transcripts and D-J rearrangement of TCR β-chain. These results suggested that, in the presence of SF, IL-11, and IL-7, primitive lymphohematopoietic progenitors differentiate toward T-cell lineage in addition to myeloid and B-cell lineages.
Collapse
|
41
|
Differentiation in Culture of Murine Primitive Lymphohematopoietic Progenitors Toward T-Cell Lineage. Blood 1999. [DOI: 10.1182/blood.v93.12.4187.412k07_4187_4195] [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
Earlier, we described a stromal cell-free two-step clonal culture system in which murine primitive lymphohematopoietic progenitors produce myeloid and B-lymphoid lineage cells. In the same culture T-cell potential of the progenitors was maintained. We now report that, in addition to myeloid and B-lymphoid cells, putative T-cell progenitors are also produced in culture. Lineage-negative (Lin−) Ly-6A/E+ c-kit+ bone marrow cells from 5-fluorouracil–treated mice were cultured in methylcellulose in the presence of SF (Steel factor), interleukin (IL)-11, and IL-7, and the resulting primary colonies were picked and pooled. When injected into severe combined immune deficiency (scid) mice, the pooled cells reconstituted the T-cell compartment of the scid mice earlier than freshly prepared primitive marrow cells. This reconstitution activity of the pooled primary colony cells was enriched in the Ly-6A/E+ and FcγRII/III−/low cell fractions. Reverse transcriptase-polymerase chain reaction (RT-PCR) and DNA-PCR analyses showed that some of the primary colony cells are differentiated sufficiently to express messenger RNA (mRNA) of T-cell receptor (TCR) β-chain and pre-TCR alpha (pT) and, although not frequently, to perform Dβ-Jβ rearrangement of the TCR gene. Micromanipulation studies confirmed the clonal origin of myeloid lineage cells and the cells positive for the T-cell–specific transcripts and D-J rearrangement of TCR β-chain. These results suggested that, in the presence of SF, IL-11, and IL-7, primitive lymphohematopoietic progenitors differentiate toward T-cell lineage in addition to myeloid and B-cell lineages.
Collapse
|
42
|
Reizis B, Leder P. Expression of the mouse pre-T cell receptor alpha gene is controlled by an upstream region containing a transcriptional enhancer. J Exp Med 1999; 189:1669-78. [PMID: 10330446 PMCID: PMC2193641 DOI: 10.1084/jem.189.10.1669] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/1999] [Indexed: 11/23/2022] Open
Abstract
The pre-T cell receptor alpha (pTalpha) protein is a critical component of the pre-T cell receptor complex in early thymocytes. The expression of the pTalpha gene is one of the earliest markers of the T cell lineage and occurs exclusively in pre-T cells. To investigate the molecular basis of thymocyte-specific gene expression, we searched for the genomic elements regulating transcription of the mouse pTalpha gene. We now report that expression of the pTalpha gene is primarily controlled by an upstream genomic region, which can drive thymocyte-specific expression of a marker gene in transgenic mice. Within this region, we have identified two specific DNase-hypersensitive sites corresponding to a proximal promoter and an upstream transcriptional enhancer. The pTalpha enhancer appears to function preferentially in pre-T cell lines and binds multiple nuclear factors, including YY1. The enhancer also contains two G-rich stretches homologous to a critical region of the thymocyte-specific lck proximal promoter. Here we show that these sites bind a common nuclear factor and identify it as the zinc finger protein ZBP-89. Our data establish a novel experimental model for thymocyte-specific gene expression and suggest an important role for ZBP-89 in T cell development.
Collapse
Affiliation(s)
- B Reizis
- Department of Genetics and the Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
43
|
Böhler T, Walcher J, Hölzl-Wenig G, Geiss M, Buchholz B, Linde R, Debatin KM. Early effects of antiretroviral combination therapy on activation, apoptosis and regeneration of T cells in HIV-1-infected children and adolescents. AIDS 1999; 13:779-89. [PMID: 10357376 DOI: 10.1097/00002030-199905070-00006] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES To assess the relationship between apoptosis, activation and regeneration of T cells, and viral replication in paediatric patients with HIV-1 infection during antiretroviral therapy (ART). DESIGN In 15 HIV-1-positive children and adolescents sequential blood samples were obtained during 16 episodes of ART using combinations of nucleosidic HIV-1 reverse transcriptase (RT)-inhibitors and HIV-1 protease inhibitors or non-nucleosidic RT-inhibitors. METHODS We assessed sensitivity of freshly isolated peripheral blood T cells towards spontaneous, anti-CD95- and anti-CD3-induced apoptosis and activation before and after 6-8 weeks of ART. Expression of CD95, CD45RA, CD45RO and CD62L on CD4 and CD8 T cells and of CD34 on mononuclear cells was studied by multiparameter flow cytometry before and after 10-12 weeks of ART. RESULTS ART caused a significant increase in absolute lymphocyte and CD4 T cell counts (P < 0.03 and P < 0.02, respectively) and a decrease in both anti-CD95- and anti-CD3-induced apoptosis of CD4 and CD8 T cells to near normal levels even in patients without complete suppression of viral replication. A significant reduction in the percentage of CD95 (but not of CD95high) CD4 T cells was observed (P < 0.005). Resting/naive cells contributed significantly (P < 0.03) to the rise in CD4 T cells especially in infants and young children. CONCLUSIONS Different mechanisms may contribute to early T cell reconstitution in HIV-1-infected children and adolescents during ART: decreased activation-induced apoptosis leading to increased survival of circulating primed/memory T cells; decreased activation-induced naive-to-memory shift increasing the frequency of circulating resting/naive T cells; increased input of haematopoietic progenitor cells from the bone marrow into the thymus and decreased intrathymic T cell death leading to an increased thymic output of naive T cells.
Collapse
Affiliation(s)
- T Böhler
- Division of Molecular Oncology, German Cancer Research Center, Heidelberg
| | | | | | | | | | | | | |
Collapse
|
44
|
Strobl H, Scheinecker C, Riedl E, Csmarits B, Bello-Fernandez C, Pickl WF, Majdic O, Knapp W. Identification of CD68+lin− Peripheral Blood Cells with Dendritic Precursor Characteristics. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.2.740] [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
Expression of CD68 (macrosialin) in the absence of surface and lysosomal lineage marker molecules is a characteristic feature of T zone-associated plasmacytoid monocytes, which were recently shown to represent precursors of dendritic cells (DC). We demonstrate here a minor population of strongly CD68-positive (CD68bright) blood cells that lack all analyzed myeloid surface (CD14−, CD33−, CD13−, CD11b−, CD11c−) and lysosomal (myeloperoxidase, MPO− and lysozyme, LZ−) marker molecules (0.4 ± 2% of the total mononuclear cells). These CD68bright, lineage marker-negative (lin−) cells can be induced to proliferate in the presence of IL-3. They do not acquire myeloid features even upon stimulation with granulocyte-macrophage CSF plus IL-1, IL-3, and IL-6. Instead, these cells develop typical DC characteristics upon culture. Furthermore, these CD68brightlin− DC precursors acquire mature DC characteristics (CD86+, CD83+, CD54bright) upon stimulation with CD40 ligand plus IL-3. A second subset of DC precursor-like blood cells was found to weakly express CD68 (0.3 ± 0.2% of the total mononuclear cells) and to coexpress several myeloid lineage associated molecules (LZ+, CD11c+, CD33+, CD13+). Cells of this second subset resemble both previously described myeloid-related peripheral blood DC and germinal center DC. Analysis of peripheral blood leukocytes for CD68 thus revealed the existence of two cell subsets that phenotypically resemble lymphoid tissue-associated DC. The unique phenotype CD68brightlin− is highly reminiscent of T zone-associated plasmacytoid monocytes. CD68brightlin− blood leukocytes also functionally resemble plasmacytoid monocytes. The lack of all analyzed myeloid features by CD68brightlin− blood leukocytes suggests that these cells arise from a novel nonmyeloid human DC differentiation pathway.
Collapse
Affiliation(s)
- Herbert Strobl
- *Institute of Immunology, Vienna International Research Cooperation Center, Novartis Forschungsinstitut
| | - Clemens Scheinecker
- *Institute of Immunology, Vienna International Research Cooperation Center, Novartis Forschungsinstitut
- ‡Department of Internal Medicine III, Division of Rheumatology, University of Vienna, Vienna, Austria
| | - Elisabeth Riedl
- *Institute of Immunology, Vienna International Research Cooperation Center, Novartis Forschungsinstitut
| | | | - Concha Bello-Fernandez
- *Institute of Immunology, Vienna International Research Cooperation Center, Novartis Forschungsinstitut
| | | | | | - Walter Knapp
- *Institute of Immunology, Vienna International Research Cooperation Center, Novartis Forschungsinstitut
- †Institute of Immunology; and
| |
Collapse
|
45
|
Barber DF, Passoni L, Wen L, Geng L, Hayday AC. Cutting Edge: The Expression In Vivo of a Second Isoform of pTα: Implications for the Mechanism of pTα Action. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.1.11] [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
A second isoform of pTα, “pTαb,” is derived from the pTα locus by tissue-specific, alternative splicing. pTαb is coexpressed in the thymus with the previously characterized form of pTα (which we term pTαa) and is also expressed in peripheral cells without pTαa. While pTαa acts to retain most TCR β-chains intracellularly, pTαb permits higher levels of cell surface TCRβ expression and facilitates signaling from a CD3-TCRβ complex.
Collapse
Affiliation(s)
| | - Lorena Passoni
- *Department of Molecular, Cell & Developmental Biology and
| | - Li Wen
- *Department of Molecular, Cell & Developmental Biology and
| | - Liping Geng
- *Department of Molecular, Cell & Developmental Biology and
| | - Adrian C. Hayday
- *Department of Molecular, Cell & Developmental Biology and
- †Section of Immunobiology, Yale University, New Haven, CT 06520
| |
Collapse
|
46
|
FLT-3 Ligand and Marrow Stroma-Derived Factors Promote CD3γ, CD3δ, CD3ζ, and RAG-2 Gene Expression in Primary Human CD34+LIN−DR− Marrow Progenitors. Blood 1998. [DOI: 10.1182/blood.v91.5.1662.1662_1662_1670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We hypothesize that early lymphoid commitment from primitive hematopoietic marrow progenitors is governed by signals from the marrow microenvironment leading to sequential induction of lineage-specific genes. Using expression of lymphoid genes as markers of differentiation, we characterize a highly purified population (>99.8% by double sorting) of primary human CD34+Lin−DR− progenitors. This population was then used to evaluate the effects of supplemental cytokines (interleukin-2 [IL-2], IL-3, IL-7, c-kit ligand), FLT-3 ligand (FL), and stroma-derived factors on lymphoid differentiation in vitro. CD3, RAG-1, Ikaros, CD10, and TdT transcripts were detected in the starting CD34+Lin−DR− population. By contrast, CD3γ, CD3δ, CD3ζ, and RAG-2 transcripts were not present in any samples tested. The presence of supplemental cytokines alone at culture initiation permitted stimulation of the expression of CD3ζ, but not of CD3γ or CD3δ. However, when FL and stroma-derived factors were added to cytokines, CD3 gene expression was induced in all samples. The predominant CD3 transcripts induced by optimal culture conditions were alternatively spliced isoforms lacking transmembrane sequences (CD3δ and CD3γ) and portions of the intracellular and extracellular domains (CD3γ). The combination of cytokines, FL, and stromal factors also provided a potent stimulus for RAG-2 gene expression. These findings show that FL in combination with stroma-derived factors provide important signals to promote early events required for lymphoid differentiation.
Collapse
|
47
|
FLT-3 Ligand and Marrow Stroma-Derived Factors Promote CD3γ, CD3δ, CD3ζ, and RAG-2 Gene Expression in Primary Human CD34+LIN−DR− Marrow Progenitors. Blood 1998. [DOI: 10.1182/blood.v91.5.1662] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
We hypothesize that early lymphoid commitment from primitive hematopoietic marrow progenitors is governed by signals from the marrow microenvironment leading to sequential induction of lineage-specific genes. Using expression of lymphoid genes as markers of differentiation, we characterize a highly purified population (>99.8% by double sorting) of primary human CD34+Lin−DR− progenitors. This population was then used to evaluate the effects of supplemental cytokines (interleukin-2 [IL-2], IL-3, IL-7, c-kit ligand), FLT-3 ligand (FL), and stroma-derived factors on lymphoid differentiation in vitro. CD3, RAG-1, Ikaros, CD10, and TdT transcripts were detected in the starting CD34+Lin−DR− population. By contrast, CD3γ, CD3δ, CD3ζ, and RAG-2 transcripts were not present in any samples tested. The presence of supplemental cytokines alone at culture initiation permitted stimulation of the expression of CD3ζ, but not of CD3γ or CD3δ. However, when FL and stroma-derived factors were added to cytokines, CD3 gene expression was induced in all samples. The predominant CD3 transcripts induced by optimal culture conditions were alternatively spliced isoforms lacking transmembrane sequences (CD3δ and CD3γ) and portions of the intracellular and extracellular domains (CD3γ). The combination of cytokines, FL, and stromal factors also provided a potent stimulus for RAG-2 gene expression. These findings show that FL in combination with stroma-derived factors provide important signals to promote early events required for lymphoid differentiation.
Collapse
|
48
|
Retrovirally Transduced CD34++ Human Cord Blood Cells Generate T Cells Expressing High Levels of the Retroviral Encoded Green Fluorescent Protein Marker In Vitro. Blood 1998. [DOI: 10.1182/blood.v91.2.431] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Human umbilical cord blood (UCB) hematopoietic stem cells (HSC) receive increased attention as a possible target for gene-transfer in gene therapy trials. Diseases affecting the lymphoid lineage, as adenosine deaminase (ADA) deficiency and acquired immunodeficiency syndrome (AIDS) could be cured by gene therapy. However, the T-cell progenitor potential of these HSC after gene-transfer is largely unknown and was up to now not testable in vitro. We show here that highly purified CD34++ Lineage marker-negative (CD34++Lin−) UCB cells generate T, natural killer (NK), and dendritic cells in a severe combined immunodeficient mouse fetal thymus organ culture (FTOC). CD34++Lin− and CD34++CD38−Lin− UCB cells express the retroviral encoded marker gene Green Fluorescent Protein (GFP) after in vitro transduction with MFG-GFP retroviral supernatant. Transduced cells were still capable of generating T, NK, and dendritic cells in the FTOC, all expressing high levels of GFP under control of the Moloney murine leukemia virus (MoMuLV) long terminal repeat promotor. We thus present an in vitro assay for thymic T-cell development out of transduced UCB HSC, using GFP as a marker gene.
Collapse
|
49
|
Retrovirally Transduced CD34++ Human Cord Blood Cells Generate T Cells Expressing High Levels of the Retroviral Encoded Green Fluorescent Protein Marker In Vitro. Blood 1998. [DOI: 10.1182/blood.v91.2.431.431_431_440] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human umbilical cord blood (UCB) hematopoietic stem cells (HSC) receive increased attention as a possible target for gene-transfer in gene therapy trials. Diseases affecting the lymphoid lineage, as adenosine deaminase (ADA) deficiency and acquired immunodeficiency syndrome (AIDS) could be cured by gene therapy. However, the T-cell progenitor potential of these HSC after gene-transfer is largely unknown and was up to now not testable in vitro. We show here that highly purified CD34++ Lineage marker-negative (CD34++Lin−) UCB cells generate T, natural killer (NK), and dendritic cells in a severe combined immunodeficient mouse fetal thymus organ culture (FTOC). CD34++Lin− and CD34++CD38−Lin− UCB cells express the retroviral encoded marker gene Green Fluorescent Protein (GFP) after in vitro transduction with MFG-GFP retroviral supernatant. Transduced cells were still capable of generating T, NK, and dendritic cells in the FTOC, all expressing high levels of GFP under control of the Moloney murine leukemia virus (MoMuLV) long terminal repeat promotor. We thus present an in vitro assay for thymic T-cell development out of transduced UCB HSC, using GFP as a marker gene.
Collapse
|
50
|
Robust, But Transient Expression of Adeno-Associated Virus-Transduced Genes During Human T Lymphopoiesis. Blood 1997. [DOI: 10.1182/blood.v90.12.4854] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractRecombinant adeno-associated viruses (rAAV) have been proposed to be gene transfer vehicles for hematopoietic stem cells with advantages over other virus-based systems due to their high titers and relative lack of dependence on cell cycle for target cell integration. We evaluated rAAV vector containing a LacZ reporter gene under the control of a cytomegalovirus (CMV) promoter in the context of primary human CD34+CD2− progenitor cells induced to undergo T-cell differentiation using an in vitro T-lymphopoiesis system. Target cells from either adult bone marrow or umbilical cord blood were efficiently transduced, and 71% to 79% CD2+ cells expressed a LacZ marker gene mRNA and produced LacZ-encoded protein after exposure to rAAV-CMV-LacZ. The impact of transgene expression on the differentiation of T cells was assessed by sequential quantitation of immunophenotypic subsets of virus-exposed cells and no alteration was noted compared with control. The durability of transgene expression was assessed and found to decay by day 35 with kinetics dependent on the multiplicity of infection. In addition, vector DNA was absent from CD4 or CD8 subselected CD3+ cells by DNA-polymerase chain reaction. These data suggest that rAAV vectors may result in robust transgene expression in primitive cells undergoing T-cell lineage commitment without toxicity or alteration in the pattern of T-cell differentiation. However, expression is transient and integration of the transgene unlikely. Recombinant AAV vectors are potentially valuable gene transfer tools for the genetic manipulation of events during T-cell ontogony but their potential in gene therapy strategies for diseases such as acquired immunodeficiency syndrome is limited.
Collapse
|