1
|
Mauvais FX, Hamel Y, Silvin A, Mulder K, Hildner K, Akyol R, Dalod M, Koumantou D, Saveanu L, Garfa M, Cagnard N, Bertocci B, Ginhoux F, van Endert P. Metallophilic marginal zone macrophages cross-prime CD8 + T cell-mediated protective immunity against blood-borne tumors. Immunity 2025; 58:843-860.e20. [PMID: 40139188 DOI: 10.1016/j.immuni.2025.02.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 10/27/2024] [Accepted: 02/28/2025] [Indexed: 03/29/2025]
Abstract
Splenic metallophilic marginal zone macrophages (MMMs) are positioned to control the dissemination of blood-borne threats. We developed a purification protocol to enable characterization of MMMs phenotypically and transcriptionally. MMM gene expression profile was enriched for pathways associated with CD8+ T cell activation and major histocompatibility complex class I (MHC class I) cross-presentation. In vitro, purified MMMs equaled conventional dendritic cells type 1 (cDC1s) in cross-priming CD8+ T cells to soluble and particulate antigens, yet MMMs employed a distinct vacuolar processing pathway. In vivo biphoton and ex vivo light-sheet imaging showed long-standing contacts with cognate T cells differentiating to effectors. MMMs cross-primed protective CD8+ T cell antitumor responses both by capturing blood-borne tumor antigens and by internalizing tumor cells seeding the spleen. This cross-priming required expression of the transcription factor Batf3 by MMMs but was independent of cDC1-mediated capture of tumor material for cross-presentation or MHC class I-dressing. Thus, MMMs combine control of the dissemination of blood-borne pathogens and tumor materials with the initiation of innate and adaptive responses.
Collapse
Affiliation(s)
- François-Xavier Mauvais
- Université Paris Cité, INSERM, CNRS, Institut Necker Enfants Malades, 75015 Paris, France; Service de Physiologie - Explorations Fonctionnelles Pédiatriques, AP-HP, Hôpital Universitaire Robert Debré, 75019 Paris, France.
| | - Yamina Hamel
- Université Paris Cité, INSERM, CNRS, Institut Necker Enfants Malades, 75015 Paris, France
| | - Aymeric Silvin
- Gustave Roussy Cancer Campus, Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Kevin Mulder
- Gustave Roussy Cancer Campus, Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Kai Hildner
- University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Medical Department 1, Deutsches Zentrum Immuntherapie, 91054 Erlangen, Germany
| | - Ramazan Akyol
- Aix Marseille Université, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Marc Dalod
- Aix Marseille Université, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Despoina Koumantou
- Université Paris Cité, Centre de recherche sur l'inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018 Paris, France; Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018 Paris, France
| | - Loredana Saveanu
- Université Paris Cité, Centre de recherche sur l'inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine site Bichat, 75018 Paris, France; Université Paris Cité, Laboratoire d'Excellence Inflamex, 75018 Paris, France
| | - Meriem Garfa
- Cell Imaging, Structure Fédérative de Recherche Necker, INSERM, US24/CNRS UMS3633, 75015 Paris, France
| | - Nicolas Cagnard
- Bioinformatics Core Facilities, Structure Fédérative de Recherche Necker, INSERM, US24/CNRS UMS3633, 75015 Paris, France
| | - Barbara Bertocci
- Université Paris Cité, INSERM, CNRS, Institut Necker Enfants Malades, 75015 Paris, France
| | - Florent Ginhoux
- Gustave Roussy Cancer Campus, Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Équipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France; Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research, (A∗STAR), Singapore, Singapore; Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Peter van Endert
- Université Paris Cité, INSERM, CNRS, Institut Necker Enfants Malades, 75015 Paris, France; Service Immunologie Biologique, AP-HP, Hôpital Universitaire Necker-Enfants Malades, 75015 Paris, France.
| |
Collapse
|
2
|
Tian L, Tomei S, Schreuder J, Weber TS, Amann-Zalcenstein D, Lin DS, Tran J, Audiger C, Chu M, Jarratt A, Willson T, Hilton A, Pang ES, Patton T, Kelly M, Su S, Gouil Q, Diakumis P, Bahlo M, Sargeant T, Kats LM, Hodgkin PD, O'Keeffe M, Ng AP, Ritchie ME, Naik SH. Clonal multi-omics reveals Bcor as a negative regulator of emergency dendritic cell development. Immunity 2021; 54:1338-1351.e9. [PMID: 33862015 DOI: 10.1016/j.immuni.2021.03.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 02/05/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022]
Abstract
Despite advances in single-cell multi-omics, a single stem or progenitor cell can only be tested once. We developed clonal multi-omics, in which daughters of a clone act as surrogates of the founder, thereby allowing multiple independent assays per clone. With SIS-seq, clonal siblings in parallel "sister" assays are examined either for gene expression by RNA sequencing (RNA-seq) or for fate in culture. We identified, and then validated using CRISPR, genes that controlled fate bias for different dendritic cell (DC) subtypes. This included Bcor as a suppressor of plasmacytoid DC (pDC) and conventional DC type 2 (cDC2) numbers during Flt3 ligand-mediated emergency DC development. We then developed SIS-skew to examine development of wild-type and Bcor-deficient siblings of the same clone in parallel. We found Bcor restricted clonal expansion, especially for cDC2s, and suppressed clonal fate potential, especially for pDCs. Therefore, SIS-seq and SIS-skew can reveal the molecular and cellular mechanisms governing clonal fate.
Collapse
Affiliation(s)
- Luyi Tian
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Sara Tomei
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Jaring Schreuder
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Tom S Weber
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Daniela Amann-Zalcenstein
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia; Single Cell Open Research Endeavour (SCORE), The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Dawn S Lin
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Jessica Tran
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Cindy Audiger
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Mathew Chu
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Andrew Jarratt
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia; Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Tracy Willson
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia; Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Adrienne Hilton
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia; Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Ee Shan Pang
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Timothy Patton
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Madison Kelly
- The Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Shian Su
- Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Quentin Gouil
- Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Peter Diakumis
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia; Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Melanie Bahlo
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia; Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Toby Sargeant
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia; Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Lev M Kats
- The Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Philip D Hodgkin
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Meredith O'Keeffe
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Ashley P Ng
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia; Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Matthew E Ritchie
- Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Shalin H Naik
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia; Single Cell Open Research Endeavour (SCORE), The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia.
| |
Collapse
|
3
|
Rodrigues PF, Tussiwand R. Novel concepts in plasmacytoid dendritic cell (pDC) development and differentiation. Mol Immunol 2020; 126:25-30. [PMID: 32739721 DOI: 10.1016/j.molimm.2020.07.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/29/2020] [Accepted: 07/02/2020] [Indexed: 01/08/2023]
Abstract
Plasmacytoid dendritic cells (pDCs) are an immune subset specialized in the production of Type I Interferons (IFNs). They are characterized by co-expression of myeloid and lymphoid markers. Their developmental origin has been studied since their discovery and the identification of a myeloid progenitor capable of generating all dendritic cell (DC) subsets, including pDCs, led to their classification within the myeloid compartment. However, recent findings challenge this hypothesis and provide evidence for a lymphoid origin for the majority of pDCs 46-48. In this review we discuss and present the original myeloid and the newer lymphoid developmental trajectories of pDCs.
Collapse
Affiliation(s)
| | - Roxane Tussiwand
- Department of Biomedicine, University of Basel, 4058, Basel, Switzerland; Laboratory of Immune Regulation, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
4
|
Tussiwand R, Gautier EL. Transcriptional Regulation of Mononuclear Phagocyte Development. Front Immunol 2015; 6:533. [PMID: 26539196 PMCID: PMC4609886 DOI: 10.3389/fimmu.2015.00533] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 10/02/2015] [Indexed: 12/23/2022] Open
Abstract
Mononuclear phagocytes (MP) are a quite unique subset of hematopoietic cells, which comprise dendritic cells (DC), monocytes as well as monocyte-derived and tissue-resident macrophages. These cells are extremely diverse with regard to their origin, their phenotype as well as their function. Developmentally, DC and monocytes are constantly replenished from a bone marrow hematopoietic progenitor. The ontogeny of macrophages is more complex and is temporally linked and specified by the organ where they reside, occurring early during embryonic or perinatal life. The functional heterogeneity of MPs is certainly a consequence of the tissue of residence and also reflects the diverse ontogeny of the subsets. In this review, we will highlight the developmental pathways of murine MP, with a particular emphasis on the transcriptional factors that regulate their development and function. Finally, we will discuss and point out open questions in the field.
Collapse
Affiliation(s)
- Roxane Tussiwand
- Department of Biomedicine, University of Basel , Basel , Switzerland
| | - Emmanuel L Gautier
- INSERM UMR_S 1166, Sorbonne Universités, UPMC Univ Paris 06, Pitié-Salpêtrière Hospital , Paris , France
| |
Collapse
|
5
|
Breton G, Lee J, Zhou YJ, Schreiber JJ, Keler T, Puhr S, Anandasabapathy N, Schlesinger S, Caskey M, Liu K, Nussenzweig MC. Circulating precursors of human CD1c+ and CD141+ dendritic cells. ACTA ACUST UNITED AC 2015; 212:401-13. [PMID: 25687281 PMCID: PMC4354370 DOI: 10.1084/jem.20141441] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Two subsets of conventional dendritic cells (cDCs) with distinct cell surface markers and functions exist in mouse and human. The two subsets of cDCs are specialized antigen-presenting cells that initiate T cell immunity and tolerance. In the mouse, a migratory cDC precursor (pre-CDC) originates from defined progenitors in the bone marrow (BM). Small numbers of short-lived pre-CDCs travel through the blood and replace cDCs in the peripheral organs, maintaining homeostasis of the highly dynamic cDC pool. However, the identity and distribution of the immediate precursor to human cDCs has not been defined. Using a tissue culture system that supports the development of human DCs, we identify a migratory precursor (hpre-CDC) that exists in human cord blood, BM, blood, and peripheral lymphoid organs. hpre-CDCs differ from premonocytes that are restricted to the BM. In contrast to earlier progenitors with greater developmental potential, the hpre-CDC is restricted to producing CD1c(+) and CD141(+) Clec9a(+) cDCs. Studies in human volunteers demonstrate that hpre-CDCs are a dynamic population that increases in response to levels of circulating Flt3L.
Collapse
Affiliation(s)
- Gaëlle Breton
- Laboratory of Molecular Immunology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065
| | - Jaeyop Lee
- Columbia University Medical Center, Department of Microbiology and Immunology, New York, NY 10032
| | - Yu Jerry Zhou
- Columbia University Medical Center, Department of Microbiology and Immunology, New York, NY 10032
| | | | | | - Sarah Puhr
- Columbia University Medical Center, Department of Microbiology and Immunology, New York, NY 10032
| | - Niroshana Anandasabapathy
- Laboratory of Molecular Immunology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065 Department of Dermatology Brigham and Women's Hospital, Boston, MA 02115
| | - Sarah Schlesinger
- Laboratory of Molecular Immunology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065
| | - Marina Caskey
- Laboratory of Molecular Immunology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065
| | - Kang Liu
- Columbia University Medical Center, Department of Microbiology and Immunology, New York, NY 10032
| | - Michel C Nussenzweig
- Laboratory of Molecular Immunology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065 Laboratory of Molecular Immunology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065
| |
Collapse
|
6
|
Xuan NT, Wang X, Nishanth G, Waisman A, Borucki K, Isermann B, Naumann M, Deckert M, Schlüter D. A20 expression in dendritic cells protects mice from LPS-induced mortality. Eur J Immunol 2014; 45:818-28. [PMID: 25472594 DOI: 10.1002/eji.201444795] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 10/22/2014] [Accepted: 11/28/2014] [Indexed: 11/10/2022]
Abstract
DCs contribute to immune homeostasis under physiological conditions and regulate the immune activation during infection. The deubiquitinase A20 inhibits the activation of NF-κB-dependent immune reactions, and prevents the hyperactivation of DCs under steady-state conditions. However, the role of DC-specific A20 under pathological conditions is unknown. Here, we demonstrate that upon injection of low-dose LPS, mice with DC-specific A20 deletion (CD11c-Cre A20(fl/fl) ) died within 6 h, whereas A20(fl/fl) controls survived. LPS-induced mortality in CD11c-Cre A20(fl/fl) mice was characterized by increased serum levels of IL-2, IL-10, IL-12, IFN-γ, and TNF. Upon LPS stimulation, the activation of NF-κB and ERK-NFATc3 pathways were enhanced in A20-deficient DCs, resulting in an increased production of IL-2, IL-12, and TNF both in vitro and in vivo. Targeted inhibition of ERK in A20-deficient DCs abolished the increased production of IL-2. A20-deficient DCs failed to induce LPS tolerance, which was independent of T cells and the intestinal flora, since T-cell depletion and decolonization of CD11c-Cre A20(fl/fl) mice could not prevent death of LPS-challenged CD11c-Cre A20(fl/fl) mice. In conclusion, these findings show that DC-specific A20 preserves immune homeostasis in steady-state conditions and is also required for LPS tolerance.
Collapse
Affiliation(s)
- Nguyen Thi Xuan
- Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Parasite fate and involvement of infected cells in the induction of CD4+ and CD8+ T cell responses to Toxoplasma gondii. PLoS Pathog 2014; 10:e1004047. [PMID: 24722202 PMCID: PMC3983043 DOI: 10.1371/journal.ppat.1004047] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 02/18/2014] [Indexed: 01/04/2023] Open
Abstract
During infection with the intracellular parasite Toxoplasma gondii, the presentation of parasite-derived antigens to CD4+ and CD8+ T cells is essential for long-term resistance to this pathogen. Fundamental questions remain regarding the roles of phagocytosis and active invasion in the events that lead to the processing and presentation of parasite antigens. To understand the most proximal events in this process, an attenuated non-replicating strain of T. gondii (the cpsII strain) was combined with a cytometry-based approach to distinguish active invasion from phagocytic uptake. In vivo studies revealed that T. gondii disproportionately infected dendritic cells and macrophages, and that infected dendritic cells and macrophages displayed an activated phenotype characterized by enhanced levels of CD86 compared to cells that had phagocytosed the parasite, thus suggesting a role for these cells in priming naïve T cells. Indeed, dendritic cells were required for optimal CD4+ and CD8+ T cell responses, and the phagocytosis of heat-killed or invasion-blocked parasites was not sufficient to induce T cell responses. Rather, the selective transfer of cpsII-infected dendritic cells or macrophages (but not those that had phagocytosed the parasite) to naïve mice potently induced CD4+ and CD8+ T cell responses, and conferred protection against challenge with virulent T. gondii. Collectively, these results point toward a critical role for actively infected host cells in initiating T. gondii-specific CD4+ and CD8+ T cell responses. CD4+ and CD8+ T cells are critical for controlling many infections. To generate a T cell response during infection, T cells must encounter the microbial peptides that they recognize bound to MHC molecules on the surfaces of other cells, such as dendritic cells. It is currently unclear how dendritic cells acquire the antigens they present to T cells during infection with many intracellular pathogens. It is possible that these antigens are phagocytosed and processed by dendritic cells, or antigens may be presented by cells that are infected by pathogens such as Toxoplasma gondii, which invades host cells independently of phagocytosis. To differentiate these pathways, we developed a novel technique to track the fate of T. gondii in vivo that distinguishes actively infected cells from those that phagocytosed parasites. This technique was used to examine each of these cell populations. We also used pharmacological inhibitors of parasite invasion, and the transfer of sort-purified infected or uninfected dendritic cells and macrophages to determine what roles phagocytosis and active invasion have in the initiation of T cell responses. Our results demonstrate that phagocytosis of parasites is not sufficient to induce CD4+ or CD8+ T cell responses, whereas infected cells are critical for this process.
Collapse
|
8
|
Saulep-Easton D, Vincent FB, Le Page M, Wei A, Ting SB, Croce CM, Tam C, Mackay F. Cytokine-driven loss of plasmacytoid dendritic cell function in chronic lymphocytic leukemia. Leukemia 2014; 28:2005-15. [PMID: 24721775 PMCID: PMC4100939 DOI: 10.1038/leu.2014.105] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 02/10/2014] [Accepted: 03/06/2014] [Indexed: 02/06/2023]
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by the accumulation of CD5+CD19+ B cells in the peripheral blood, and in primary and secondary lymphoid organs. A major complication associated with CLL is severe recurrent infections, which are often fatal. Vulnerability to infection is due to a wide variety of immunological defects, yet the initiating events of immunodeficiency in CLL are unclear. Using CLL patient samples and a mouse model of CLL, we have discovered that plasmacytoid dendritic cells (pDCs), which underpin the activity of effector immune cells critical for anti-viral immunity and anti-tumor responses, are reduced in number and functionally impaired in progressive CLL. As a result, the levels of interferon alpha (IFNα) production, a cytokine critical for immunity, are markedly reduced. Lower pDC numbers with impaired IFNα production was due to the decreased expression of FMS-like tyrosine kinase 3 receptor (Flt3) and Toll-like receptor 9 (TLR9), respectively. Reduced Flt3 expression was reversed using inhibitors of TGF-β and TNF, an effect correlating with a reduction in tumor load. Defects in pDC numbers and function offer a new insight into mechanisms underpinning the profound immunodeficiency affecting CLL patients and provide a potentially novel avenue for restoring immuno-competency in CLL.
Collapse
Affiliation(s)
- D Saulep-Easton
- Department of Immunology, Monash University Central Clinical School, Alfred Medical Research and Education Precinct (AMREP), 89 Commercial Road, Melbourne, Victoria, Australia
| | - F B Vincent
- Department of Immunology, Monash University Central Clinical School, Alfred Medical Research and Education Precinct (AMREP), 89 Commercial Road, Melbourne, Victoria, Australia
| | - M Le Page
- Department of Immunology, Monash University Central Clinical School, Alfred Medical Research and Education Precinct (AMREP), 89 Commercial Road, Melbourne, Victoria, Australia
| | - A Wei
- 1] Department of Haematology, The Alfred Hospital, 55 Commercial Road, Melbourne, Victoria, Australia [2] Australian Centre for Blood Diseases, Division of Blood Cancers, Monash University Central Clinical School, Alfred Medical Research and Education Precinct (AMREP), 89 Commercial Road, Melbourne, Victoria, Australia
| | - S B Ting
- 1] Department of Haematology, The Alfred Hospital, 55 Commercial Road, Melbourne, Victoria, Australia [2] Australian Centre for Blood Diseases, Division of Blood Cancers, Monash University Central Clinical School, Alfred Medical Research and Education Precinct (AMREP), 89 Commercial Road, Melbourne, Victoria, Australia
| | - C M Croce
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, 460 West 12th Avenue, Columbus, OH, USA
| | - C Tam
- Department of Haematology, Peter MacCallum Cancer Centre, St. Andrews's Place, East Melbourne, Victoria, Australia
| | - F Mackay
- Department of Immunology, Monash University Central Clinical School, Alfred Medical Research and Education Precinct (AMREP), 89 Commercial Road, Melbourne, Victoria, Australia
| |
Collapse
|
9
|
Abstract
Dendritic cells (DCs) drive both adaptive and innate immunity. Recent findings support the notion that distinct subsets of classical DCs favor alternative modules of immunity, acting on innate lymphoid-like cells (ILCs) and T cells similarly to promote either ILC1/Th1/CTL- or ILC3/Th17-type responses. Coordination between DC subsets and their favored immune module might imply that the genetic programs for DC diversification preceded the emergence of recombination-activating gene-dependent adaptive immunity and operate initially in coordinating ILC repertoires for appropriate responses against pathogens. Consequently, understanding the molecular basis of DC developmental and diversification is important for an underlying appreciation of immune regulation. Currently, the basis for DC development into the recognized subsets/lineages is only partially understood, based on the requirements for several transcription factors including PU.1, Bcl11a, Irf8, E2-2, Id2, Irf4, Irf8, Batf3, and other BATF family members. This chapter will briefly review recent transcriptional aspects of DC development and function and then highlight some currently unresolved questions.
Collapse
Affiliation(s)
- Kenneth M Murphy
- School of Medicine, Department of Pathology and Immunology, Washington University, St. Louis, Missouri, USA; School of Medicine, Howard Hughes Medical Institute, Washington University, St. Louis, Missouri, USA.
| |
Collapse
|
10
|
Satpathy AT, Wu X, Albring JC, Murphy KM. Re(de)fining the dendritic cell lineage. Nat Immunol 2012; 13:1145-54. [PMID: 23160217 DOI: 10.1038/ni.2467] [Citation(s) in RCA: 333] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 09/27/2012] [Indexed: 12/12/2022]
Abstract
Dendritic cells (DCs) are essential mediators of innate and adaptive immune responses. Study of these critical cells has been complicated by their similarity to other hematopoietic lineages, particularly monocytes and macrophages. Progress has been made in three critical areas of DC biology: the characterization of lineage-restricted progenitors in the bone marrow, the identification of cytokines and transcription factors required during differentiation, and the development of genetic tools for the visualization and depletion of DCs in vivo. Collectively, these advances have clarified the nature of the DC lineage and have provided novel insights into their function during health and disease.
Collapse
Affiliation(s)
- Ansuman T Satpathy
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | | | | | | |
Collapse
|
11
|
John B, Ricart B, Tait Wojno ED, Harris TH, Randall LM, Christian DA, Gregg B, De Almeida DM, Weninger W, Hammer DA, Hunter CA. Analysis of behavior and trafficking of dendritic cells within the brain during toxoplasmic encephalitis. PLoS Pathog 2011; 7:e1002246. [PMID: 21949652 PMCID: PMC3174247 DOI: 10.1371/journal.ppat.1002246] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 07/20/2011] [Indexed: 12/25/2022] Open
Abstract
Under normal conditions the immune system has limited access to the brain; however, during toxoplasmic encephalitis (TE), large numbers of T cells and APCs accumulate within this site. A combination of real time imaging, transgenic reporter mice, and recombinant parasites allowed a comprehensive analysis of CD11c+ cells during TE. These studies reveal that the CNS CD11c+ cells consist of a mixture of microglia and dendritic cells (DCs) with distinct behavior associated with their ability to interact with parasites or effector T cells. The CNS DCs upregulated several chemokine receptors during TE, but none of these individual receptors tested was required for migration of DCs into the brain. However, this process was pertussis toxin sensitive and dependent on the integrin LFA-1, suggesting that the synergistic effect of signaling through multiple chemokine receptors, possibly leading to changes in the affinity of LFA-1, is involved in the recruitment/retention of DCs to the CNS and thus provides new insights into how the immune system accesses this unique site. Toxoplasmic encephalitis (TE), caused by the protozoan parasite Toxoplasma gondii, can be potentially life threatening especially in immuno-compromised individuals. Immune cells including dendritic cells have been shown to accumulate in the brain during chronic toxoplasmosis; however, little is known about their function, their behavior in vivo, and the mechanisms by which they migrate into the brain. In the present studies, we utilize a combination of real time imaging, transgenic reporter mice, and recombinant parasites to reveal the distinct behavior and morphologies of dendritic cells within the brain and their ability to interact with parasites and effector T cells during TE. The CNS DCs were also found to exhibit a unique chemokine receptor expression pattern during infection, and the migration of DCs into the brain was mediated through a pertussis toxin (which blocks signaling downstream of several chemokine receptors) sensitive process and dependent on the integrin LFA-1. There is currently a poor understanding of the events that lead to DC recruitment to the CNS during inflammation in general, and our studies provide new insights into the mechanisms by which antigen-presenting cells gain access to the brain during infection.
Collapse
Affiliation(s)
- Beena John
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Brendon Ricart
- Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Elia D. Tait Wojno
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Tajie H. Harris
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Louise M. Randall
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - David A. Christian
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Beth Gregg
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Daniel Manzoni De Almeida
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Wolfgang Weninger
- The Centenary Institute for Cancer Medicine and Cell Biology, Newtown, Australia
| | - Daniel A. Hammer
- Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
12
|
Satpathy AT, Murphy KM, KC W. Transcription factor networks in dendritic cell development. Semin Immunol 2011; 23:388-97. [PMID: 21924924 PMCID: PMC4010935 DOI: 10.1016/j.smim.2011.08.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Accepted: 08/19/2011] [Indexed: 12/23/2022]
Abstract
Dendritic cells (DCs) are a heterogeneous population within the mononuclear phagocyte system (MPS) that derive from bone marrow precursors. Commitment and specification of hematopoietic progenitors to the DC lineage is critical for the proper induction of both immunity and tolerance. This review summarizes the important cytokines and transcription factors required for differentiation of the DC lineage as well as further diversification into specific DC subsets. We highlight recent advances in the characterization of immediate DC precursors arising from the common myeloid progenitor (CMP). Particular emphasis is placed on the corresponding temporal expression of relevant factors involved in regulating developmental options.
Collapse
Affiliation(s)
- Ansuman T Satpathy
- Department of Pathology and Immunology, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | | | | |
Collapse
|
13
|
Abstract
BACKGROUND/AIMS We have compared dendritic cell (DC) function derived from the alcoholic liver disease (ALD) sensitive Long-Evans (LE) and resistant Fischer rat strains to determine if the influence of ethanol on DCs was dependent on ALD. METHODS The LE and Fischer rats were fed an ethanol-containing or isocaloric control liquid diet for 8 weeks and comparisons were made to LE rats injected with thioacetamide as a liver disease control. DCs were isolated from the spleen after expansion with human Fms-like tyrosine kinase receptor 3 ligand plasmid. Maturation markers CD86, CD80, CD40 and MHC-II were analysed by flow cytometry with or without lipopolysaccharide and poly I:C stimulation. Production of tumour necrosis factor-α (TNF-α), interferon-γ (IFN-γ), interleukin (IL)-12p40 and IL-10 cytokines and the antigen presentation ability of DCs was determined. RESULTS Only LE rats developed ALD characterized by liver injury, elevated alanine aminotransferase levels and steatosis; CD86 and CD40 expression was decreased in LE but not Fischer rats. Reduced TNF-α, IFN-γ, IL-12, proinflammatory and enhanced IL-10 cytokine production was found in DCs isolated from ethanol-fed LE but not Fischer rats. Allostimulatory activity was reduced in LE compared with the Fischer strain. In contrast, DCs isolated from thioacetamide-induced liver damage displayed a reduction only in IL-12p40; TNF-α, IL-10 and IFN-α production as well as antigen presenting ability remained intact compared with controls. CONCLUSIONS ALD sensitive LE rats exhibited characteristics of a suppressed DC phenotype that was not observed following thioacetamide-induced liver disease, which suggests an important role for ALD in altering the host cellular and humoral immune responses.
Collapse
Affiliation(s)
- Dechun Feng
- The Department of Medicine, Liver Research Center, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence, RI 02905, USA
| | | | | | | |
Collapse
|
14
|
Atibalentja DF, Murphy KM, Unanue ER. Functional redundancy between thymic CD8α+ and Sirpα+ conventional dendritic cells in presentation of blood-derived lysozyme by MHC class II proteins. THE JOURNAL OF IMMUNOLOGY 2010; 186:1421-31. [PMID: 21178002 DOI: 10.4049/jimmunol.1002587] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We evaluated the presentation of blood-derived protein Ags by APCs in the thymus. Two conventional dendritic cells (cDCs), the CD8α(+)Sirpα(-)CD11c(hi) (CD8α(+) cDC) and the CD8α(-)Sirpα(+)CD11c(hi) (Sirpα(+) cDC), were previously identified as presenting MHC class II bound peptides from hen egg white lysozyme (HEL) injected intravenously. All thymic APCs acquired the injected HEL, with the plasmacytoid dendritic cell being the best, followed by the Sirpα(+) cDC and the CD8α(+) cDC. Both cDCs induced to similar extent negative selection and regulatory T cells in HEL TCR transgenic mice, indicating a redundant role of the two cDC subsets in the presentation of blood-borne HEL. Immature dendritic cells or plasmacytoid dendritic cells were considerably less efficient. Batf3(-/-) mice, with significantly reduced numbers of CD8α(+) cDCs, were not impaired in HEL presentation by I-A(k) molecules of thymic APCs. Lastly, clodronate liposome treatment of TCR transgenic mice depleted blood APCs including Sirpα(+) cDCs without affecting the number of thymic APCs. In such treated mice, there was no effect on negative selection or regulatory T cells in mice when administering HEL, indicating that the T cell responses were mediated primarily by the cDCs localized in the thymus.
Collapse
Affiliation(s)
- Danielle F Atibalentja
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | |
Collapse
|
15
|
Reboulet RA, Hennies CM, Garcia Z, Nierkens S, Janssen EM. Prolonged antigen storage endows merocytic dendritic cells with enhanced capacity to prime anti-tumor responses in tumor-bearing mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 185:3337-47. [PMID: 20720209 PMCID: PMC3021914 DOI: 10.4049/jimmunol.1001619] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tumor cell vaccination with irradiated autologous tumor cells is a promising approach to activate tumor-specific T cell responses without the need for tumor Ag identification. However, uptake of dying cells by dendritic cells (DCs) is generally a noninflammatory or tolerizing event to prevent the development of autoreactive immune responses. In this study, we describe the mechanisms that confer the potent T cell priming capacity of a recently identified a population of DCs (merocytic DCs [mcDCs]) that potently primes both CD8(+) and CD4(+) T cells to cell-associated Ags upon uptake of apoptotic cells. mcDCs acquired cell-associated materials through a process of merocytosis that is defined by the uptake of small particles that are stored in nonacidic compartments for prolonged periods, sustained Ag presentation, and the induction of type I IFN. T cells primed by mcDCs to cell-associated Ags exhibit increased primary expansion, enhanced effector function, and increased memory formation. By using transgenic T cell transfer models and endogenous models, we show that treatment of tumor-bearing mice with mcDCs that have been exposed to dying tumor cells results in tumor suppression and increased host survival through the activation of naive tumor-specific CD8(+) T cells as well as the reinvigoration of tumor-specific T cells that had been rendered nonresponsive by the tumor in vivo. The potent capacity of mcDCs to prime both CD4(+) and CD8(+) T cells to cell-associated Ags under immunosuppressive conditions makes this DC subset an attractive target for tumor therapies as well as interventional strategies for autoimmunity and transplantation.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Neoplasm/metabolism
- Antigens, Neoplasm/physiology
- CD11b Antigen/biosynthesis
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/pathology
- Cell Line, Tumor
- Cell Proliferation
- Cell Survival/immunology
- Cross-Priming/immunology
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/pathology
- Dose-Response Relationship, Immunologic
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/pathology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/prevention & control
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Molecular Sequence Data
Collapse
Affiliation(s)
- Rachel A Reboulet
- Division of Molecular Immunology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229, USA
| | | | | | | | | |
Collapse
|
16
|
Bankoti J, Burnett A, Navarro S, Miller AK, Rase B, Shepherd DM. Effects of TCDD on the fate of naive dendritic cells. Toxicol Sci 2010; 115:422-34. [PMID: 20211938 PMCID: PMC2871756 DOI: 10.1093/toxsci/kfq063] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Accepted: 02/09/2010] [Indexed: 12/21/2022] Open
Abstract
The environmental contaminant, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), causes immune suppression via activation of the aryl hydrocarbon receptor. Dendritic cells (DCs), the professional antigen-presenting cells in the immune system, are adversely affected by TCDD. We hypothesized that TCDD alters DC homeostasis, resulting in a loss of DCs in naive mice. To test this hypothesis, C57Bl/6 mice were gavaged with either vehicle or an immunosuppressive dose of TCDD (15 microg/kg). TCDD exposure decreased the frequency and number of splenic CD11c(high) DCs on day 7 when compared with vehicle-treated controls. TCDD increased the expression of CD86 and CD54, while decreasing the frequency of splenic CD11c(high) DCs expressing CD11a and major histocompatibility complex (MHC) class II. Moreover, TCDD selectively decreased the CD11c(high)CD8alpha(-)33D1(+) splenic DCs specialized at activating CD4(+) T cells but did not affect the regulatory CD11c(high)CD8alpha(+)DEC205(+) splenic DCs. TCDD did not alter the number or frequency of CD11c(low) splenic DCs but decreased their MHC class II and CD11a expression. Loss of splenic CD11c(high) DCs was independent of Fas-mediated apoptosis and was not due to alterations in the numbers of common DC precursors in the bone marrow or their ability to generate steady-state DCs in vitro. Instead, increased CCR7 expression on CD11c(high) DCs suggested involvement of a migratory event. Popliteal and brachial lymph node CD11c(+) cells showed elevated levels of MHC class II and CD40 following TCDD exposure. Collectively, this study shows the presence of a TCDD-sensitive splenic DC subpopulation in naive mice, suggesting that TCDD may induce suppression of T-cell-mediated immunity by disrupting DC homeostasis.
Collapse
Affiliation(s)
- Jaishree Bankoti
- Department of Biomedical and Pharmaceutical Sciences
- Center for Environmental Health Sciences, University of Montana, Missoula, Montana 59812
| | - Andrea Burnett
- Center for Environmental Health Sciences, University of Montana, Missoula, Montana 59812
| | - Severine Navarro
- Center for Environmental Health Sciences, University of Montana, Missoula, Montana 59812
| | - Andrea K. Miller
- Center for Environmental Health Sciences, University of Montana, Missoula, Montana 59812
| | - Ben Rase
- Center for Environmental Health Sciences, University of Montana, Missoula, Montana 59812
| | - David M. Shepherd
- Department of Biomedical and Pharmaceutical Sciences
- Center for Environmental Health Sciences, University of Montana, Missoula, Montana 59812
| |
Collapse
|
17
|
Garnache-Ottou F, Feuillard J, Ferrand C, Biichle S, Trimoreau F, Seilles E, Salaun V, Garand R, Lepelley P, Maynadié M, Kuhlein E, Deconinck E, Daliphard S, Chaperot L, Beseggio L, Foisseaud V, Macintyre E, Bene MC, Saas P, Jacob MC. Extended diagnostic criteria for plasmacytoid dendritic cell leukaemia. Br J Haematol 2009; 145:624-36. [PMID: 19388928 DOI: 10.1111/j.1365-2141.2009.07679.x] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The diagnosis of plasmacytoid dendritic cell leukaemia (pDCL) is based on the immunophenotypic profile: CD4(+) CD56(+) lineage(neg) CD45RA(+)/RO(neg) CD11c(neg) CD116(low) CD123(+) CD34(neg) CD36(+) HLA-DR(+). Several studies have reported pDCL cases that do not express this exact profile or expressing some lineage antigens that could thus be misdiagnosed. This study aimed to validate pDCL-specific markers for diagnosis by flow-cytometry or quantitative reverse transcription polymerase chain reaction on bone marrow samples. Expression of markers previously found in normal pDC was analysed in 16 pDCL, four pDCL presenting an atypical phenotype (apDCL) and 113 non-pDC - lymphoid or myeloid - acute leukaemia. CD123 was expressed at significantly higher levels in pDCL and apDCL. BDCA-2 was expressed on 12/16 pDCL and on 2/4 apDCL, but was never detected in the 113 non-pDC acute leukaemia cases. BDCA-4 expression was found on 13/16 pDCL, but also in 12% of non-pDC acute leukaemia. High levels of LILRA4 and TCL1A transcripts distinguished pDCL and apDCL from all other acute leukaemia (except B-cell acute lymphoblastic leukaemia for TCL1A). We thus propose a diagnosis strategy, scoring first the CD4(+) CD56(+/-) MPO(neg) cCD3(neg) cCD79a(neg) CD11c(neg) profile and then the CD123(high), BDCA-2 and BDCA-4 expression. Atypical pDCL can be also identified this way and non-pDC acute leukaemia excluded: this scoring strategy is useful for diagnosing pDCL and apDCL.
Collapse
Affiliation(s)
- Francine Garnache-Ottou
- INSERM UMR645, Université of Franche-Comté, Etablissement Français du Sang Bourgogne Franche-Comté, 1 boulevard A. Fleming, Besançon, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Birnberg T, Bar-On L, Sapoznikov A, Caton ML, Cervantes-Barragán L, Makia D, Krauthgamer R, Brenner O, Ludewig B, Brockschnieder D, Riethmacher D, Reizis B, Jung S. Lack of conventional dendritic cells is compatible with normal development and T cell homeostasis, but causes myeloid proliferative syndrome. Immunity 2008; 29:986-97. [PMID: 19062318 DOI: 10.1016/j.immuni.2008.10.012] [Citation(s) in RCA: 188] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2008] [Revised: 07/31/2008] [Accepted: 10/13/2008] [Indexed: 12/23/2022]
Abstract
Dendritic cells are critically involved in the promotion and regulation of T cell responses. Here, we report a mouse strain that lacks conventional CD11c(hi) dendritic cells (cDCs) because of constitutive cell-type specific expression of a suicide gene. As expected, cDC-less mice failed to mount effective T cell responses resulting in impaired viral clearance. In contrast, neither thymic negative selection nor T regulatory cell generation or T cell homeostasis were markedly affected. Unexpectedly, cDC-less mice developed a progressive myeloproliferative disorder characterized by prominent extramedullary hematopoiesis and increased serum amounts of the cytokine Flt3 ligand. Our data identify a critical role of cDCs in the control of steady-state hematopoiesis, revealing a feedback loop that links peripheral cDCs to myelogenesis through soluble growth factors, such as Flt3 ligand.
Collapse
Affiliation(s)
- Tal Birnberg
- Department of Immunology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Martin AP, Rankin S, Pitchford S, Charo IF, Furtado GC, Lira SA. Increased expression of CCL2 in insulin-producing cells of transgenic mice promotes mobilization of myeloid cells from the bone marrow, marked insulitis, and diabetes. Diabetes 2008; 57:3025-33. [PMID: 18633103 PMCID: PMC2570399 DOI: 10.2337/db08-0625] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To define the mechanisms underlying the accumulation of monocytes/macrophages in the islets of Langerhans. RESEARCH DESIGN AND METHODS We tested the hypothesis that macrophage accumulation into the islets is caused by overexpression of the chemokine CCL2. To test this hypothesis, we generated transgenic mice and evaluated the cellular composition of the islets by immunohistochemistry and flow cytometry. We determined serum levels of CCL2 by enzyme-linked immunosorbent assay, determined numbers of circulating monocytes, and tested whether CCL2 could mobilize monocytes from the bone marrow directly. We examined development of diabetes over time and tested whether CCL2 effects could be eliminated by deletion of its receptor, CCR2. RESULTS Expression of CCL2 by beta-cells was associated with increased numbers of monocytes in circulation and accumulation of macrophages in the islets of transgenic mice. These changes were promoted by combined actions of CCL2 at the level of the bone marrow and the islets and were not seen in animals in which the CCL2 receptor (CCR2) was inactivated. Mice expressing higher levels of CCL2 in the islets developed diabetes spontaneously. The development of diabetes was correlated with the accumulation of large numbers of monocytes in the islets and did not depend on T- and B-cells. Diabetes could also be induced in normoglycemic mice expressing low levels of CCL2 by increasing the number of circulating myeloid cells. CONCLUSIONS These results indicate that CCL2 promotes monocyte recruitment by acting both locally and remotely and that expression of CCL2 by insulin-producing cells can lead to insulitis and islet destruction.
Collapse
Affiliation(s)
- Andrea P Martin
- Immunology Institute, Mount Sinai School of Medicine, New York, New York, USA
| | | | | | | | | | | |
Collapse
|
20
|
Abstract
Hematopoietic stem cell transplantation is an important experimental tool and therapeutic modality. Its efficacy and toxicity are both linked to a GvH reaction that is initiated by donor T cells recognizing recipient APC, of which DC are the most potent. In most tissues recipient DC are replaced after transplantation because they turnover rapidly from BM-derived precursors. However, in a number of sites, notably the skin, recipient DC may persist and even self-renew for many months after transplantation. Understanding the homeostasis of different APC populations and how they are related to the induction of alloreactivity may help to improve the therapeutic benefit of transplantation.
Collapse
Affiliation(s)
- M P Collin
- Department of Gene and Cell Medicine, Mount Sinai Medical School, New York, New York 10029, USA.
| | | | | |
Collapse
|
21
|
Mielcarek M, Kucera KA, Nash R, Torok-Storb B, McKenna HJ. Identification and characterization of canine dendritic cells generated in vivo. Biol Blood Marrow Transplant 2007; 13:1286-93. [PMID: 17950915 PMCID: PMC2185715 DOI: 10.1016/j.bbmt.2007.07.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2007] [Accepted: 07/16/2007] [Indexed: 10/22/2022]
Abstract
Emerging evidence suggests that host dendritic cells (DC) initiate and regulate graft-versus-host and graft-versus-tumor reactions after allogeneic hematopoietic cell transplantation (HCT). Even though decades of experimentation in the preclinical canine HCT model have substantially improved our understanding of the biology and safety of HCT in human patients, the in vivo phenotype of potent antigen-presenting cells in dogs is poorly defined. Therefore, peripheral blood leukocytes were obtained from dogs treated with recombinant human Flt3-ligand and phenotypically distinct cell populations, including putative DC, were purified by 4-color flow-cytometry and tested for their stimulatory potential in allogeneic mixed lymphocyte cultures (MLC). Cells characterized by surface expression of CD11c and HLA-DR, and absence of expression of CD14 and DM5, a marker of mature granulocytes, were found to be highly potent stimulators in allogeneic MLC. In contrast, all other immunophenotypically different cell populations tested had either weak or absent allostimulatory potential. Transmission electron microscopy of CD11c+/HLA-DR+/CD14-/DM5- cells revealed the morphology similar to that described for DC in humans and ex vivo-generated canine DC, including long cytoplasmic extensions, discrete lysosomes, and an abundant Golgi apparatus and endoplasmatic reticulum. In summary, CD11c+/HLA-DR+/CD14-/DM5- cells obtained from canine peripheral blood have functional and morphologic characteristics similar to those of human myeloid DC.
Collapse
Affiliation(s)
- Marco Mielcarek
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109-1024, USA.
| | | | | | | | | |
Collapse
|
22
|
Matthews K, Rhind SM, Gossner AG, Dalziel RG, Hopkins J. The effects of gene gun delivered pIL-3 adjuvant on skin pathology and cytokine expression. Vet Immunol Immunopathol 2007; 119:233-42. [PMID: 17628699 DOI: 10.1016/j.vetimm.2007.05.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Revised: 05/16/2007] [Accepted: 05/29/2007] [Indexed: 11/18/2022]
Abstract
The aim of this study was to investigate skin immunopathology following gene gun delivery of plasmid-encoding interleukin 3 (pIL-3) and hence explore the possible mechanisms of its adjuvant activity. Using the sheep as the experimental model, expressible pIL-3 was administered to the epidermis and the dermal/epidermal junction and its effects on the skin were assessed by histopathology, immunohistology and quantitative RT-PCR for a range of pro-inflammatory and immune response polarizing cytokines. Delivery of both functional and non-functional plasmids caused an acute inflammatory response with the infiltration of neutrophils and micro-abscess formation; however, the response to pIL-3 was more severe and was also associated with an early (24 h) infiltration of B cells and a later accumulation of CD172a-/CD45RA+ dendritic cells (DC). In terms of cytokine transcript expression, an early TNFalpha response was stimulated by gene gun delivery of plasmid-associated gold beads, which coincided with an immediate infiltration of neutrophils. However, only pIL-3 triggered the short-lived expression of IL-3 (peaking at 6 h) and significant long-term increases in both TNFalpha and IL-1beta. pIL-3 did not affect the expression of the immune response polarizing cytokines, IL-10 and IL-12.
Collapse
Affiliation(s)
- K Matthews
- Centre of Infectious Diseases, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Summerhall, Edinburgh EH9 1QH, UK
| | | | | | | | | |
Collapse
|
23
|
Sailaja G, Skountzou I, Quan FS, Compans RW, Kang SM. Human immunodeficiency virus-like particles activate multiple types of immune cells. Virology 2007; 362:331-41. [PMID: 17276476 PMCID: PMC1974898 DOI: 10.1016/j.virol.2006.12.014] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2006] [Revised: 11/09/2006] [Accepted: 12/08/2006] [Indexed: 12/28/2022]
Abstract
The rapid spread of human immunodeficiency virus (HIV) worldwide makes it a high priority to develop an effective vaccine. Since live attenuated or inactivated HIV is not likely to be approved as a vaccine due to safety concerns, HIV virus like particles (VLPs) offer an attractive alternative because they are safe due to the lack of a viral genome. Although HIV VLPs have been shown to induce humoral and cellular immune responses, it is important to understand the mechanisms by which they induce such responses and to improve their immunogenicity. We generated HIV VLPs, and VLPs containing Flt3 ligand (FL), a dendritic cell growth factor, to target VLPs to dendritic cells, and investigated the roles of these VLPs in the initiation of adaptive immune responses in vitro and in vivo. We found that HIV-1 VLPs induced maturation of dendritic cells and monocyte/macrophage populations in vitro and in vivo, with enhanced expression of maturation markers and cytokines. Dendritic cells pulsed with VLPs induced activation of splenocytes resulting in increased production of cytokines. VLPs containing FL were found to increase dendritic cells and monocyte/macrophage populations in the spleen when administered to mice. Administration of VLPs induced acute activation of multiple types of cells including T and B cells as indicated by enhanced expression of the early activation marker CD69 and down-regulation of the homing receptor CD62L. VLPs containing FL were an effective form of antigen in activating immune cells via dendritic cells, and immunization with HIV VLPs containing FL resulted in enhanced T helper type 2-like immune responses.
Collapse
MESH Headings
- AIDS Vaccines/immunology
- Animals
- Antigens, CD/biosynthesis
- Antigens, Differentiation, T-Lymphocyte/biosynthesis
- Cell Proliferation
- Cells, Cultured
- Cytokines/biosynthesis
- Dendritic Cells/immunology
- Flow Cytometry
- Gene Products, env/genetics
- Gene Products, env/immunology
- Gene Products, gag/genetics
- Genes, env
- HIV Antibodies/blood
- HIV Antigens/genetics
- HIV Antigens/immunology
- HIV-1/genetics
- HIV-1/immunology
- Humans
- L-Selectin/biosynthesis
- Lectins, C-Type
- Lymphocyte Subsets/immunology
- Macrophages/immunology
- Membrane Proteins/immunology
- Mice
- Mice, Inbred BALB C
- Models, Animal
- Spleen/cytology
- Spleen/immunology
- Spodoptera/cytology
- Vaccines, Virosome/immunology
- Virosomes/immunology
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/immunology
Collapse
Affiliation(s)
- Gangadhara Sailaja
- Department of Microbiology and Immunology, Emory Vaccine Center Emory University School of Medicine, 1510 Clifton Rd, Atlanta, GA 30322
| | - Ioanna Skountzou
- Department of Microbiology and Immunology, Emory Vaccine Center Emory University School of Medicine, 1510 Clifton Rd, Atlanta, GA 30322
| | - Fu-Shi Quan
- Department of Microbiology and Immunology, Emory Vaccine Center Emory University School of Medicine, 1510 Clifton Rd, Atlanta, GA 30322
| | - Richard W. Compans
- Department of Microbiology and Immunology, Emory Vaccine Center Emory University School of Medicine, 1510 Clifton Rd, Atlanta, GA 30322
| | - Sang-Moo Kang
- Department of Microbiology and Immunology, Emory Vaccine Center Emory University School of Medicine, 1510 Clifton Rd, Atlanta, GA 30322
| |
Collapse
|
24
|
Paczesny S, Li YP, Li N, Latger-Cannard V, Marchal L, Ou-Yang JP, Bordigoni P, Stoltz JF, Eljaafari A. Efficient generation of CD34+ progenitor-derived dendritic cells from G-CSF-mobilized peripheral mononuclear cells does not require hematopoietic stem cell enrichment. J Leukoc Biol 2007; 81:957-67. [PMID: 17229904 DOI: 10.1189/jlb.0406296] [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: 11/24/2022] Open
Abstract
As a result of their potent antigen-presentation function, dendritic cells (DC) are important tools for cell therapy programs. In vitro-generated DC from enriched CD34+ hematopoietic stem cells (HSC; enriched CD34 DC) have already proven their efficiency in Phase I/II clinical trials. Here, we investigated whether enrichment of CD34+ HSC before the onset of culture was absolutely required for their differentiation into DC. With this aim, we developed a new two-step culture method. PBMC harvested from G-CSF-mobilized, healthy patients were expanded for 7 days during the first step, with early acting cytokines, such as stem cell factor, fetal liver tyrosine kinase 3 ligand (Flt-3L), and thrombopoietin. During the second step, expanded cells were then induced to differentiate into mature DC in the presence of GM-CSF, Flt-3L, and TNF-alpha for 8 days, followed by LPS exposure for 2 additional days. Our results showed that the rate of CD34+/CD38+/lineageneg cells increased 19.5+/-10-fold (mean+/-sd) during the first step, and the expression of CD14, CD1a, CD86, CD80, and CD83 molecules was up-regulated markedly following the second step. When compared with DC generated from enriched CD34+ cells, which were expanded for 7 days before differentiation, DC derived from nonenriched peripheral blood stem cells showed a similar phenotye but higher yields of production. Accordingly, the allogeneic stimulatory capacity of the two-step-cultured DC was as at least as efficient as that of enriched CD34 DC. In conclusion, we report herein a new two-step culture method that leads to high yields of mature DC without any need of CD34+ HSC enrichment.
Collapse
Affiliation(s)
- Sophie Paczesny
- Hematology Department, Children's Hospital, CHU Nancy, France, and Department of Pediatrics, University of Michigan Cancer Center, 1500 East Medical Center Drive, Ann Arbor, MI 48109-0942, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Zhang YL, Chen SS, Yang KG, Su L, Deng YC, Liu CZ. Functional expression, purification, and characterization of human Flt3 ligand in the Pichia pastoris system. Protein Expr Purif 2006; 42:246-54. [PMID: 15914030 DOI: 10.1016/j.pep.2005.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2004] [Revised: 03/31/2005] [Accepted: 04/01/2005] [Indexed: 11/30/2022]
Abstract
Flt3 ligand (FL) is a potent hematopoietic cytokine that affects the growth and differentiation of hematopoietic progenitor and stem cells both in vivo and in vitro. Pichia pastoris transformants secreting high-level rhFL were obtained using 'yeastern blotting' method and the expression level in liquid was about 30 mg/L. rhFL was purified to about 95% purity with overnight dialysis, filtration and an anion-exchange step. Further purification steps employing Sephacryl S-200 and reverse-phase HPLC raised the purity to over 99%. The purified rhFL possessed correct N-terminal amino acid sequence and positive Western blotting bands. SDS-PAGE and mass spectrometry analysis showed molecular weight of rhFL was about 21 and 34 kDa, suggesting that rhFL was glycosylated. The result of capillary electrophoresis showed that its pI is 3.12-4.72. Endo H deglycosylation analysis indicated that there was O-glycosylation besides N-glycosylation in rhFL secreted from P. pastoris. Bioactivity assay showed that the purified rhFL had dose-dependent expansion activity on bone marrow nucleated cells.
Collapse
Affiliation(s)
- Yan-Li Zhang
- National Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China.
| | | | | | | | | | | |
Collapse
|
26
|
Sheng JR, Li L, Ganesh BB, Vasu C, Prabhakar BS, Meriggioli MN. Suppression of Experimental Autoimmune Myasthenia Gravis by Granulocyte-Macrophage Colony-Stimulating Factor Is Associated with an Expansion of FoxP3+Regulatory T Cells. THE JOURNAL OF IMMUNOLOGY 2006; 177:5296-306. [PMID: 17015715 DOI: 10.4049/jimmunol.177.8.5296] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Dendritic cells (DCs) have the potential to activate or tolerize T cells in an Ag-specific manner. Although the precise mechanism that determines whether DCs exhibit tolerogenic or immunogenic functions has not been precisely elucidated, growing evidence suggests that DC function is largely dependent on differentiation status, which can be manipulated using various growth factors. In this study, we investigated the effects of mobilization of specific DC subsets-using GM-CSF and fms-like tyrosine kinase receptor 3-ligand (Flt3-L)-on the susceptibility to induction of experimental autoimmune myasthenia gravis (EAMG). We administered GM-CSF or Flt3-L to C57BL/6 mice before immunization with acetylcholine receptor (AChR) and observed the effect on the frequency and severity of EAMG development. Compared with AChR-immunized controls, mice treated with Flt3-L before immunization developed EAMG at an accelerated pace initially, but disease frequency and severity was comparable at the end of the observation period. In contrast, GM-CSF administered before immunization exerted a sustained suppressive effect against the induction of EAMG. This suppression was associated with lowered serum autoantibody levels, reduced T cell proliferative responses to AChR, and an expansion in the population of FoxP3+ regulatory T cells. These results highlight the potential of manipulating DCs to expand regulatory T cells for the control of autoimmune diseases such as MG.
Collapse
MESH Headings
- Animals
- Autoimmune Diseases/therapy
- Cell Communication/immunology
- Cell Proliferation/drug effects
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Forkhead Transcription Factors
- Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage
- Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology
- Immunization
- Membrane Proteins/administration & dosage
- Membrane Proteins/pharmacology
- Mice
- Mice, Inbred C57BL
- Myasthenia Gravis, Autoimmune, Experimental/immunology
- Myasthenia Gravis, Autoimmune, Experimental/prevention & control
- Myasthenia Gravis, Autoimmune, Experimental/therapy
- Receptors, Cholinergic/administration & dosage
- Receptors, Cholinergic/immunology
- T-Lymphocytes, Regulatory/cytology
Collapse
Affiliation(s)
- Jian Rong Sheng
- Department of Neurology and Rehabilitation, University of Illinois, Chicago, IL 60612, USA
| | | | | | | | | | | |
Collapse
|
27
|
Ceredig R, Rauch M, Balciunaite G, Rolink AG. Increasing Flt3L availability alters composition of a novel bone marrow lymphoid progenitor compartment. Blood 2006; 108:1216-22. [PMID: 16675711 DOI: 10.1182/blood-2005-10-006643] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Abstract
We have recently described a CD19– B220+CD117low bone marrow subpopulation with B, T, and myeloid developmental potential, which we have called “early progenitors with lymphoid and myeloid potential” or EPLM. These cells also expressed Fms-like tyrosine kinase 3, Flt3, or CD135. Treatment of mice with the corresponding ligand, Flt3L, showed a 50-fold increase in EPLM. In addition to the expected increase in dendritic cell numbers, Flt3L treatment had a reversible inhibitory effect on B lymphopoiesis. Limiting dilution analysis of sorted EPLM from Flt3L-treated mice showed that B-lymphocyte progenitor activity was reduced 20-fold, but that myeloid and T-cell progenitor activity was largely preserved. EPLM from treated mice transiently reconstituted the thymus and bone marrow of recipient mice, generating cohorts of functional T and B cells in peripheral lymphoid organs. Thus, Flt3L treatment results in a dramatic increase in a novel bone marrow cell with lymphoid and myeloid progenitor activity.
Collapse
Affiliation(s)
- Rhodri Ceredig
- Department of Clinical and Biological Sciences, University of Basel, Mattenstrasse 28, CH-4058 Basel, Switzerland
| | | | | | | |
Collapse
|
28
|
Abstract
Dendritic cells (DC) develop in vivo from hematopoietic precursor cells. This process can be mimicked in vitro by growth factor stimulation. Among those factors granulocyte-macrophage colony-stimulating factor (GM-CSF) is the best known and most widely used for generation of rodent and human DC of the myeloid lineage. GM-CSF is often combined with interleukin-4 (IL-4) to suppress macrophage (Mph) outgrowth in cultures of human cells, but this does not apply to the mouse, and detailed analyses on the role of IL-4 are rare. Despite evidence for the importance of GM-CSF for DC development derived from in vitro data, GM-CSF-deficient mice are largely normal with respect to their DC populations. This raised the interest in other growth factors for DC. IL-3 can also support DC growth in vitro, but has been neglected for some years. Now it has been revived by a series of publications. In this review, some new features of myeloid DC regarding their early developmental stages, the GM-CSF/IL-4-interplay, and the role of IL-3 are summarized.
Collapse
Affiliation(s)
- Manfred B Lutz
- Department of Dermatology, University Hospital Erlangen, Hartmannstr. 14, 91052 Erlangen, Germany.
| |
Collapse
|
29
|
Abstract
"Nature has provided, in the white corpuscles as you call them-in the phagocytes as we call them-a natural means of devouring and destroying all disease germs. There is at bottom only one genuinely scientific treatment for all diseases, and that is to stimulate the phagocytes." So opined B.B. in G.B. Shaw's The Doctor's Dilemma in a dramatic restatement of a key portion of Ilya Metchnikoff's Nobel Prize address: "Whenever the organism enjoys immunity, the introduction of infectious microbes is followed by the accumulation of mobile cells, of white corpuscles of the blood in particular which absorb the microbes and destroy them. The white corpuscles and the other cells capable of doing this have been designated 'phagocytes,' (i.e., devouring cells) and the whole function that ensures immunity has been given the name of 'phagocytosis'". Based on these insights into the foundation of resistance to infectious disease, Metchnikoff was awarded the 1908 Nobel Prize in Physiology or Medicine together with Paul Ehrlich (Fig. 1). Although both were cited for discoveries in immunity, the contributions of the two men seem worlds apart. Ehrlich's studies did not deal with generic responses to infection, but rather with the highly specific nature of antibodies and their relationship to the cells producing them: "As the cell receptor is obviously preformed, and the artificially produced antitoxin only the consequence, i.e. secondary, one can hardly fail to assume that the antitoxin is nothing else but discharged components of the cell, namely receptors discharged in excess". But biological systems are just that-systems-and the parts need to work together. And so we arrive, a century later, at an appreciation for just how intimately related these two seemingly disparate aspects of host defense really are.
Collapse
Affiliation(s)
- Ronald N Germain
- Lymphocyte Biology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA.
| |
Collapse
|
30
|
Edwan JH, Perry G, Talmadge JE, Agrawal DK. Flt-3 ligand reverses late allergic response and airway hyper-responsiveness in a mouse model of allergic inflammation. THE JOURNAL OF IMMUNOLOGY 2004; 172:5016-23. [PMID: 15067083 DOI: 10.4049/jimmunol.172.8.5016] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Flt3 ligand (Flt3-L) is a growth factor for dendritic cells and induces type 1 T cell responses. We recently reported that Flt3-L prevented OVA-induced allergic airway inflammation and suppressed late allergic response and airway hyper-responsiveness (AHR). In the present study we examined whether Flt3-L reversed allergic airway inflammation in an established model of asthma. BALB/c mice were sensitized and challenged with OVA, and AHR to methacholine was established. Then mice with AHR were randomized and treated with PBS or 6 microg of Flt3-L i.p. for 10 days. Pulmonary functions and AHR to methacholine were examined after rechallenge with OVA. Treatment with Flt3-L of presensitized mice significantly suppressed (p < 0.001) the late allergic response, AHR, bronchoalveolar lavage fluid total cellularity, absolute eosinophil counts, and inflammation in the lung tissue. There was a significant decrease in proinflammatory cytokines (TNF-alpha, IL-4, and IL-5) in bronchoalveolar lavage fluid, with a significant increase in serum IL-12 and a decrease in serum IL-5 levels. There was no significant effect of Flt3-L treatment on serum IL-4 and serum total IgE levels. Sensitization with OVA significantly increased CD11b(+)CD11c(+) cells in the lung, and this phenomenon was not significantly affected by Flt3-L treatment. These data suggest that Flt3-L can reverse allergic airway inflammation and associated changes in pulmonary functions in murine asthma model.
Collapse
Affiliation(s)
- Jehad H Edwan
- Department of Medical Microbiology and Immunology, and Center for Allergy, Asthma, and Immunology, Creighton University School of Medicine, Omaha, NE 68178, USA
| | | | | | | |
Collapse
|
31
|
Sevilla N, McGavern DB, Teng C, Kunz S, Oldstone MBA. Viral targeting of hematopoietic progenitors and inhibition of DC maturation as a dual strategy for immune subversion. J Clin Invest 2004; 113:737-45. [PMID: 14991072 PMCID: PMC351322 DOI: 10.1172/jci20243] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2003] [Accepted: 12/02/2003] [Indexed: 11/17/2022] Open
Abstract
DCs play a pivotal role in bringing forth innate and adaptive immune responses. Viruses can specifically target DCs, rendering them ineffective in stimulating T cells, which can ultimately lead to immunosuppression. In the present study we have identified several potential mechanisms by which lymphocytic choriomeningitis virus (LCMV) induces immunosuppression in its natural murine host. The immunosuppressive LCMV variant clone 13 (Cl 13) infects DCs and interferes with their maturation and antigen-presenting capacity as evidenced by a significant reduction in the surface expression of MHC class I, MHC class II, CD40, CD80, and CD86 molecules. Additionally, Cl 13 infects hematopoietic progenitor cells both in vivo and in vitro, impairing their development. One mechanism by which hematopoietic progenitors are developmentally impaired is through the Cl 13-induced production of IFN-alpha and IFN-beta (IFN-alpha/beta). Mice deficient in the receptor for IFN-alpha/beta show a normal differentiation of progenitors into DCs despite viral infection. Thus, a virus can evolve a strategy to boost its survival by preventing the maturation of DCs from infected progenitor cells and by reducing the expression of antigen-presenting and costimulatory molecules on developed DCs.
Collapse
Affiliation(s)
- Noemí Sevilla
- The Scripps Research Institute, Division of Virology, Department of Neuropharmacology, La Jolla, California, USA.
| | | | | | | | | |
Collapse
|
32
|
Masten BJ, Olson GK, Kusewitt DF, Lipscomb MF. Flt3 Ligand Preferentially Increases the Number of Functionally Active Myeloid Dendritic Cells in the Lungs of Mice. THE JOURNAL OF IMMUNOLOGY 2004; 172:4077-83. [PMID: 15034019 DOI: 10.4049/jimmunol.172.7.4077] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In the present study, we investigated the effects of in vivo Flt3L administration on the generation, phenotype, and function of lung dendritic cells (DCs) to evaluate whether Flt3L favors the expansion and maturation of a particular DC subset. Injection of Flt3L into mice resulted in an increased number of CD11c-expressing lung DCs, preferentially in the alveolar septa. FACS analysis allowed us to quantify a 19-fold increase in the absolute numbers of CD11c-positive, CD45R/B220 negative DCs in the lungs of Flt3L-treated mice over vehicle-treated mice. Further analysis revealed a 90-fold increase in the absolute number of myeloid DCs (CD11c positive, CD45R/B220 negative, and CD11b positive) and only a 3-fold increase of lymphoid DCs (CD11c positive, CD45R/B220 negative, and CD11b negative) from the lungs of Flt3L-treated mice over vehicle-treated mice. Flt3L-treated lung DCs were more mature than vehicle-treated lung DCs as demonstrated by a significantly higher percentage of cells expressing MHC class II, CD86, and CD40. Freshly isolated Flt3L lung DCs were not fully mature, because after an overnight culture they continued to increase accessory molecule expression. Functionally, Flt3L-treated lung DCs were more efficient than vehicle-treated DCs at stimulating naive T cell proliferation. Our data show that administration of Flt3L favors the expansion of myeloid lung DCs over lymphoid DCs and enhanced their ability to stimulate naive lymphocytes.
Collapse
Affiliation(s)
- Barbara J Masten
- Department of Pathology, University of New Mexico, Albuquerque, NM 87131, USA.
| | | | | | | |
Collapse
|
33
|
Morin J, Faideau B, Gagnerault MC, Lepault F, Boitard C, Boudaly S. Passive transfer of flt-3L-derived dendritic cells delays diabetes development in NOD mice and associates with early production of interleukin (IL)-4 and IL-10 in the spleen of recipient mice. Clin Exp Immunol 2004; 134:388-95. [PMID: 14632742 PMCID: PMC1808900 DOI: 10.1111/j.1365-2249.2003.02308.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
CD11c+/CD11b+dendritic cells (DC) with high levels of major histocompatibility complex (MHC) class II and co-stimulatory molecules have been derived from spleen cells cultured with granulocyte-macrophage colony stimulating factor (GM-CSF) + flt-3L + interleukin (IL)-6 (flt-3L-DC). Investigating in vivo the function of DC in non-obese diabetic mice (NOD), we showed that a single injection of this in vitro-derived subset of DC prevents the development of diabetes into prediabetic female mice. In contrast, DC derived from bone marrow cells cultured with GM-CSF + IL-4 [bone marrow (BM)-DC] induced no protection. Moreover, protection against diabetes following injection of flt-3L-DC was associated with IL-4 and IL-10 production in the spleen and the pancreatic lymph nodes of recipient mice, indicating that this DC population is able to polarize the immune response towards a Th2 pathway. As we shown previously, NOD BM-DC exhibit an enhanced capacity to produce IL-12p70 in response to lipopolysaccharide (LPS) and anti-CD40 stimulation compared to BM-DC from control mice. In contrast, NOD flt-3L-DC, as their control mouse counterpart, produced no IL-12p70 to these stimuli. Our findings show that a subset of DC, characterized by a mature phenotype and the absence of IL-12p70 production can be derived from NOD mouse spleen favouring IL-4 and IL-10 regulatory responses and protection from diabetes development.
Collapse
Affiliation(s)
- J Morin
- Laboratoire d'Immunologie, Génétique et Traitement des Maladies Métaboliques et du Diabète, Inserm U 561, Hôpital Saint-Vincent-de-Paul, Paris, France
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
Hematopoietic stem cell transplants (SCT) are used in the treatment of neoplastic diseases, in addition to congenital, autoimmune, and inflammatory disorders. Both autologous and allogeneic SCT are used, depending on donor availability and the type of disease being treated, resulting in different morbidity and outcomes. In both types of SCT, immune regulation via graft manipulation is being studied, although with highly different targeted outcomes. In general, autologous SCT have lower treatment-related morbidity and mortality, but a higher incidence of tumor relapse, and graft manipulation targets immune augmentation and/or the reduction of immune tolerance. In contrast, allogeneic SCT have a higher incidence of treatment-related morbidity and mortality and a significantly longer time of disease progression, and the targeted outcomes or graft manipulation focus on a reduction in graft versus host disease (GVHD). One source of the increased relapse rate and shorter overall survival (OS) following high dose chemotherapy (HDT) and autologous SCT is the immune tolerance that limits host response, both innate and antigen (Ag) specific, against the tumor. The immune tolerance that is observed is due in part to the tumor burden and prior cytotoxic therapy. Therefore, graft manipulation, as an adjuvant therapeutic approach in autologous SCT, is primarily focused on non-specific or specific immune augmentation using cytokines and vaccines. Recently, manipulation of the infused product as a form of cellular therapy has begun to also focus on approaches to reduce immune tolerance found in transplant patients, both prior to and following HDT and SCT. To this end, graft manipulation to reduce the presence of Fas Ligand (FasL)-expressing cells or interleukin (IL)10 and tumor growth factor (TGF)beta production has been proposed. In contrast to autologous transplantation, graft manipulation during allogeneic transplantation is used extensively. This includes limiting the infusion of T cells within the product or as a donor leukocyte infusion (DLI), resulting in a reduction in GVHD and the induction of long-term survivors. Indeed, allogeneic SCT provide the only curative therapy for patients with chronic myelogenous leukemia (CML), refractory acute leukemia, and myelodysplasia. The curative potential of allogeneic SCT is reduced, however, by the development of GVHD, a potentially lethal T-cell-mediated immune response targeting host tissues [Int. Arch. Allergy Immunol. 102 (1993) 309, J. Exp. Med. 183 (1996) 589]. The morbidity and mortality associated with GVHD limit this technology, resulting focus on those patients who have no alternative therapeutic options or who have advanced disease. Thus, allogeneic SCT provide one of the few statistically supported demonstrations of therapeutic efficacy by T cells (comparison of allogeneic to autologous transplantation). In contrast to autologous transplantation, control of GVHD following allogeneic SCT focuses on immune suppression and the induction of tolerance. Here too, graft manipulation is appropriate, and there are numerous studies of T-cell depletion to reduce GVHD, with or without the isolation and infusion of T cells as DLI. Additional strategies are examining the isolation and infusion of T cells with graft versus leukemia (GVL) activity to reduce GVHD and/or the infusion of genetically manipulated and/or selected cellular populations (monocytes or dendritic cells (DC)) to induce tolerance. Therefore, depending upon the type of transplant, the goals associated with graft manipulation can be radically different. In this review, we emphasize using graft manipulation to regulate immune tolerance and anergy in association with SCT. Although this paper focuses on hematopoietic SCT, it should be noted that these strategies are relevant to conditions other than neoplastic and congenital diseases, including solid organ transplants, and autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- James E Talmadge
- Nebraska Medical Center, University of Nebraska Medical Center 987660, Omaha, NE 68198-7660, USA.
| |
Collapse
|
35
|
Gitlitz BJ, Figlin RA, Kiertscher SM, Moldawer N, Rosen F, Roth MD. Phase I trial of granulocyte macrophage-colony stimulating factor and interleukin-4 as a combined immunotherapy for patients with cancer. J Immunother 2003; 26:171-8. [PMID: 12616109 DOI: 10.1097/00002371-200303000-00010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Antigen-presenting cells (APC), such as dendritic cells (DC), are the key component of many cancer immunotherapy strategies. However, DCs comprise a rare population of clinically obtainable cells and are compromised in function in cancer-bearing hosts. Clinical trials therefore rely upon DC generated ex vivo. The authors hypothesized that systemic administration of granulocyte-macrophage colony-stimulating factor (GM-CSF) plus interleukin (IL)-4 might lead to the differentiation of DC from their precursors and enhance their number and function in vivo, as it does in vitro. Subjects with advanced malignancies were treated in this phase I, multiple cohort, dose-escalation trial combining GM-CSF (2.5 microgram/kg/d) plus IL-4 (0-6.0 microgram/kg/d). A cycle consisted of 14 days of cytokine therapy and 14 days of observation (cohorts A-D), or alternating 7-day treatment and observation periods (cohort E). Subjects were followed clinically to determine a maximally tolerated dose (MTD), and complimentary in vitro studies were performed to determine a biologically active dose (BAD). Twenty-one subjects received treatment on this outpatient-based protocol. Treatment was well tolerated and generally characterized by Grade 1 and 2 cytokine related toxicities. The MTD was determined to be GM-CSF 2.5 microgram/kg/d plus IL-4 6.0 microgram/kg/d (cohort E). Treatment in cohort D (GM-CSF 2.5 microgram/kg/d plus IL-4 4.0 microgram/kg/d) was well tolerated and resulted in a BAD. Systemic GM-CSF plus IL-4 provides a mechanism for increasing the number and function of APC in cancer patients. Future clinical applications of this strategy are numerous and include the potential as a strong vaccine adjuvant.
Collapse
Affiliation(s)
- Barbara J Gitlitz
- Division of Hematology/Oncology, Jonsson Comprehensive Cancer Center, UCLA School of Medicine, Los Angeles, USA
| | | | | | | | | | | |
Collapse
|
36
|
Evans TG, Hasan M, Galibert L, Caron D. The use of Flt3 ligand as an adjuvant for hepatitis B vaccination of healthy adults. Vaccine 2002; 21:322-9. [PMID: 12450708 DOI: 10.1016/s0264-410x(02)00454-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A phase I/II clinical trial was carried out to determine the safety of Flt3 ligand used as a vaccine adjuvant when administered to healthy human volunteers on two different schedules. In the first phase of this study, Flt3 ligand was administered SQ at a dose of 20 microg/kg (to a maximum of 1500 microg) every day (N=10) or every other day (N=10) for 1 week. The Flt3 ligand injection series was followed 1 day later by the first of three vaccinations with the licensed hepatitis B vaccine. In the second phase of the trial, 30 volunteers received either Flt3 ligand or placebo on the alternate day schedule in a randomized, double-blind design. The Flt3 ligand injections were safe and very well-tolerated. The number of lineage negative, HLA-DR(hi), CD11c(+), CD123(-) dendritic cells (DCs) increased 23-fold, and the lineage negative, HLA-DR(hi), CD11c(-), CD(123 bright) pre-DCs increased 6-fold. There was an associated increase in monocytes and WBCs in the Flt3 ligand recipients. Despite the marked increase in peripheral circulating dendritic cells, no increase was observed in the hepatitis B antibody titers induced after vaccination.
Collapse
Affiliation(s)
- Thomas G Evans
- Division of Infectious Diseases, University of California, Davis, PSSB, Suite 500, 4150 V St, Sacramento, CA 95817, USA.
| | | | | | | |
Collapse
|
37
|
Mobilization of dendritic cell precursors in patients with cancer by flt3 ligand allows the generation of higher yields of cultured dendritic cells. J Immunother 2002. [PMID: 12000870 DOI: 10.1097/00002371-200205000-00011] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Flt3 ligand (Flt3L) stimulates the proliferation and differentiation of hematopoietic cells. Subcutaneous Flt3L administration has been shown to effectively manage some murine cancers and in humans, to lead to an increase in peripheral blood monocyte and dendritic cell (DC) counts. In the current study, we determined the effects of Flt3L therapy on patients with melanoma and renal cancer, and in particular, if Flt3L could be used either by enhancing the immunization of patients with melanoma to tumor antigen peptides in vivo, or by mobilizing DC precursors to allow the production of larger numbers of cultured DC. Flt3 ligand administration resulted in a 19-fold increase in DC counts in the peripheral blood of patients. The DC generated in vivo appeared only partially activated, expressing increased levels of CD86, CD33, and major histocompatibility complex class II, but no or low levels of CD80 and CD83. This partial activation may account for the lack of enhanced immune responses to melanoma antigens and absence of clinical responses in the patients even in combination with antigen immunization. Flt3 ligand administration did result, however, in a 7-fold increased yield of monocytes per liter of blood from leukapheresed patients. Dendritic cells were as readily generated from monocytes collected before and after Flt3L therapy, and they stimulated allogeneic T-cell proliferation in a mixed leukocyte reaction to a similar magnitude. Thus, the use of Flt3L may be an important method to mobilize DC precursors to allow patient therapy with larger numbers of cultured DC.
Collapse
|
38
|
Miller G, Pillarisetty VG, Shah AB, Lahrs S, Xing Z, DeMatteo RP. Endogenous granulocyte-macrophage colony-stimulating factor overexpression in vivo results in the long-term recruitment of a distinct dendritic cell population with enhanced immunostimulatory function. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:2875-85. [PMID: 12218100 DOI: 10.4049/jimmunol.169.6.2875] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
GM-CSF is critical for dendritic cell (DC) survival and differentiation in vitro. To study its effect on DC development and function in vivo, we used a gene transfer vector to transiently overexpress GM-CSF in mice. We found that up to 24% of splenocytes became CD11c+ and the number of DC increased up to 260-fold to 3 x 10(8) cells. DC numbers remained substantially elevated even 75 days after treatment. The DC population was either CD8alpha+CD4- or CD8alpha-CD4- but not CD8alpha+CD4+ or CD8alpha-CD4+. This differs substantially from subsets recruited in normal or Flt3 ligand-treated mice or using GM-CSF protein injections. GM-CSF-recruited DC secreted extremely high levels of TNF-alpha compared with minimal amounts in DC from normal or Flt3 ligand-treated mice. Recruited DC also produced elevated levels of IL-6 but almost no IFN-gamma. GM-CSF DC had robust immune function compared with controls. They had an increased rate of Ag capture and caused greater allogeneic and Ag-specific T cell stimulation. Furthermore, GM-CSF-recruited DC increased NK cell lytic activity after coculture. The enhanced T cell and NK cell immunostimulation by GM-CSF DC was in part dependent on their secretion of TNF-alpha. Our findings show that GM-CSF can have an important role in DC development and recruitment in vivo and has potential application to immunotherapy in recruiting massive numbers of DC with enhanced ability to activate effector cells.
Collapse
MESH Headings
- Adenoviridae/genetics
- Adenoviridae/immunology
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/physiology
- Animals
- Antigens/metabolism
- Cell Line
- Cell Movement/genetics
- Cell Movement/immunology
- Coculture Techniques
- Colorectal Neoplasms/metabolism
- Colorectal Neoplasms/prevention & control
- Cytotoxicity, Immunologic/genetics
- Dendritic Cells/classification
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/transplantation
- Genetic Vectors/administration & dosage
- Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis
- Granulocyte-Macrophage Colony-Stimulating Factor/blood
- Granulocyte-Macrophage Colony-Stimulating Factor/genetics
- Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
- Humans
- Immunophenotyping
- Injections, Intravenous
- Killer Cells, Natural/immunology
- Lymphocyte Activation/genetics
- Lymphocyte Activation/immunology
- Male
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/prevention & control
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Spleen/anatomy & histology
- Spleen/cytology
- Spleen/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Cells, Cultured/transplantation
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/physiology
Collapse
Affiliation(s)
- George Miller
- Hepatobiliary Service, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | | | | | | | | | | |
Collapse
|
39
|
Pisarev VM, Parajuli P, Mosley RL, Chavez J, Zimmerman D, Winship D, Talmadge JE. Flt3 ligand and conjugation to IL-1beta peptide as adjuvants for a type 1, T-cell response to an HIV p17 gag vaccine. Vaccine 2002; 20:2358-68. [PMID: 12009292 DOI: 10.1016/s0264-410x(02)00096-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The adjuvant activity of Flt3 ligand (Flt3L) and conjugation to an interleukin (IL)-1beta bioactive fragment were compared, either alone or in combination, for their ability to induce T- and B-cell responses to the HGP-30 peptide sequence (amino acids 86-115 of human immunodeficiency virus (HIV) gag p17). The efficiency of HGP-30/IL-1beta conjugation, Flt3L administration or both as adjuvants was examined and all were found to augment similar levels of delayed type hypersensitivity (DTH) responses. In contrast, significant antigen (Ag)-specific types 1 and 2 T-cell ELISPOT responses were induced only by the combination of adjuvants. Further, in vitro sensitization with HGP-30 selectively increased Ag-specific, type 1 T-cell and cytotoxic T lymphocyte (CTL) responses to HGP-30-derived nonapeptide epitopes, while type 2 responses declined as measured in the ELISPOT assay. No serum antibodies to HGP-30 were induced unless HGP-30 was conjugated to keyhole-limpet hemocyanin. This suggests that a combination adjuvant strategy using Flt3L and conjugation to a biologically active IL-1beta fragment may be used to preferentially increase type 1 T-cell and CTL responses to HIV-1 gag antigenic epitopes.
Collapse
Affiliation(s)
- Vladimir M Pisarev
- Laboratory of Transplantation Immunology, Department of Pathology/Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-7660, USA.
| | | | | | | | | | | | | |
Collapse
|
40
|
Jackson SH, Alicea C, Owens JW, Eigsti CL, Malech HL. Characterization of an early dendritic cell precursor derived from murine lineage-negative hematopoietic progenitor cells. Exp Hematol 2002; 30:430-9. [PMID: 12031649 DOI: 10.1016/s0301-472x(02)00792-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVE We define characteristics of a dendritic cell (DC) precursor generated from murine lineage-negative (Lin(-)) Sca1(+) hematopoietic progenitor cells (HPC). MATERIALS AND METHODS Lin(-)Sca1(+) HPC cultured 9 days in 100 ng/mL stem cell factor (SCF), 20 ng/mL interleukin-3 (IL-3), 50 ng/mL monocyte colony-stimulating factor (M-CSF), 5 ng/mL granulocyte-monocyte colony-stimulating factor (GM-CSF), and 25 ng/mL FLT3-ligand (FLT3-L) proliferate 387-fold and differentiate into DC precursors. Switch to > or =100 ng/mL GM-CSF + 1500 U/mL IL-4 or 500 U/mL tumor necrosis factor-alpha (TNF-alpha) for 3 days induces development into immature DC that are responsive to bacterial lipopolysaccharide (LPS)-induced maturation. RESULTS Lin(-)Sca1(+) HPC in the first 9 days of culture differentiate into DC precursors expressing surface CD11b(bright), CD11c(mod), CD86(low-mod), major histocompatibility class II antigen (MHC) II(low), DEC 205(low), but are surface CD40(-) and contain high levels of intracellular MHC II. Unlike immature DC described by others, these DC precursors are refractory to maturation with LPS and minimally stimulate allogeneic T lymphocytes in mixed leukocyte reactions (MLR). Switch to high-dose GM-CSF alone with IL-4 or TNF-alpha differentiates these DC precursors into immature DC. LPS treatment of the immature DC results in mature DC that express surface CD40(high) and CD86(high), secrete IL-1beta and IL-12, and strongly stimulate MLR. CONCLUSIONS These studies define a distinct DC precursor derived from murine HPC that precedes development of immature and mature DC.
Collapse
Affiliation(s)
- Sharon H Jackson
- The Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892-1886, USA.
| | | | | | | | | |
Collapse
|
41
|
Kwon TK, Park JW. Intramuscular co-injection of naked DNA encoding HBV core antigen and Flt3 ligand suppresses anti-HBc antibody response. Immunol Lett 2002; 81:229-34. [PMID: 11947930 DOI: 10.1016/s0165-2478(02)00039-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Flt3 ligand, a recently described growth factor affecting early hematopoietic progenitor cells, can also support the expansion of dendritic cells secreting IL-12. Its potential use in a clinical setting has been suggested. Here, we studied the effect of in situ delivery of Flt3 ligand plasmid (FL) on the antibody response induced by DNA vaccine encoding wild-type hepatitis B virus core antigen (HBc/w). Intramuscular injection of FL increased the expression of DEC205 and the size of splenocytes, and immunization with HBc/w or HBc/w-transfected EL-4 cells induced strong anti-HBc antibody responses in mice. However, intramuscular injection of FL with HBc/w significantly suppressed HBc/w-induced antibody response in a dose-dependent manner. Suppression of immune response by FL injection was the most prominent when FL and HBc/w were co-injected at the same time and the same site. These results suggest that FL may inhibit humoral response induced by DNA-type vaccination, and DC locally expanded by FL may not have proper functions for induction of humoral response.
Collapse
Affiliation(s)
- Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, 194 Dong-San Dong, Jung-Gu, Taegu 700-712, Republic of Korea
| | | |
Collapse
|
42
|
Rini BI, Paintal A, Vogelzang NJ, Gajewski TF, Stadler WM. Flt-3 ligand and sequential FL/interleukin-2 in patients with metastatic renal carcinoma: clinical and biologic activity. J Immunother 2002; 25:269-77. [PMID: 12000869 DOI: 10.1097/00002371-200205000-00010] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Flt3 (fms-like tyrosine kinase 3) ligand, a cytokine that stimulates increases in the number of dendritic cells (DC) in vivo, has been shown to have antitumor activity in murine models via an immune-mediated mechanism. Therefore, we examined the clinical activity of this cytokine in patients with an immunologic-responsive cancer, metastatic renal cell carcinoma. Flt3 ligand (25 microg/kg subcutaneous) was administered daily for the first 14 days of a 28-day cycle. Although the treatment was well tolerated and was confirmed to induce expansion of lineage (Lin)-/HLA-DR+/CD11c+ myeloid DC and Lin-/HLA-DR+/CD123+ plasmacytoid DC, no clinical activity was observed. Reasoning that DC expanded by Flt3 ligand might potentiate the clinical activity of low-dose interleukin-2, a second study was conducted of sequential administration of 25 microg/kg of Flt3 ligand daily for 7 days was followed by 11 x 10(6) IU of subcutaneous interleukin-2 for 4 consecutive days x 4 weeks. In this study, increased numbers of circulating DC were again observed, which was followed by increased numbers of activated T cells, confirming a biologic effect of each cytokine. However, toxicity and clinical efficacy were similar to what has been seen with low-dose interleukin-2 alone, with two minor responses observed. These results demonstrate that Flt3 ligand, although capable of inducing expansion of circulating myeloid and plasmacytoid DC in patients with metastatic renal cell carcinoma, lacks significant clinical activity at the doses and schedules examined.
Collapse
Affiliation(s)
- Brian I Rini
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, 5841 S Maryland Avenue, Chicago, IL 60637, U.S.A
| | | | | | | | | |
Collapse
|
43
|
De Smedt T, Smith J, Baum P, Fanslow W, Butz E, Maliszewski C. Ox40 costimulation enhances the development of T cell responses induced by dendritic cells in vivo. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:661-70. [PMID: 11777959 DOI: 10.4049/jimmunol.168.2.661] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Dendritic cells (DCs) are bone marrow-derived APCs that display unique properties aimed at stimulating naive T cells. Several members of the TNF/TNFR families have been implicated in T cell functions. In this study, we examined the role that Ox40 costimulation might play on the ability of DCs to regulate CD4(+) and CD8(+) T cell responses in vivo. Administration of anti-mouse Ox40 mAb enhanced the Th response induced by immunization with Ag-pulsed DCs, and introduced a bias toward a Th1 immune response. However, anti-Ox40 treatment enhanced the production of Th2 cytokines in IFN-gamma(-/-) mice after immunization with Ag-pulsed DCs, suggesting that the production of IFN-gamma during the immune response could interfere with the development of Th2 lymphocytes induced by DCs. Coadministration of anti-Ox40 with DCs during Ag rechallenge enhanced both Th1 and Th2 responses induced during a primary immunization with DCs, and did not reverse an existing Th2 response. This suggests that Ox40 costimulation amplifies an ongoing immune response, regardless of Th differentiation potential. In an OVA-TCR class II-restricted adoptive transfer system, anti-Ox40 treatment greatly enhanced the level of cytokine secretion per Ag-specific CD4(+) T cell induced by immunization with DCs. In an OVA-TCR class I-restricted adoptive transfer system, administration of anti-Ox40 strongly enhanced expansion, IFN-gamma secretion, and cytotoxic activity of Ag-specific CD8(+) T cells induced by immunization with DCs. Thus, by enhancing immune responses induced by DCs in vivo, the Ox40 pathway might be a target for immune intervention in therapeutic settings that use DCs as Ag-delivery vehicles.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adoptive Transfer
- Animals
- Antibodies, Monoclonal/administration & dosage
- CD4-Positive T-Lymphocytes/cytology
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- Cell Movement/immunology
- Cytokines/biosynthesis
- Dendritic Cells/immunology
- Dendritic Cells/transplantation
- Emulsions
- Female
- Hemocyanins/administration & dosage
- Injections, Intravenous
- Injections, Subcutaneous
- Interferon-gamma/deficiency
- Interferon-gamma/genetics
- Lymphocyte Activation/genetics
- Lymphocyte Activation/immunology
- Lymphocyte Count
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Mice, Transgenic
- Receptors, Antigen, T-Cell/administration & dosage
- Receptors, Antigen, T-Cell/deficiency
- Receptors, Antigen, T-Cell/genetics
- Receptors, OX40
- Receptors, Tumor Necrosis Factor
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/transplantation
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Th2 Cells/immunology
- Th2 Cells/metabolism
- Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
- Tumor Necrosis Factor Receptor Superfamily, Member 7/physiology
Collapse
Affiliation(s)
- Thibaut De Smedt
- Discovery Research Department, Immunex Corporation, Seattle, WA 98101, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Pawlowska AB, Hashino S, McKenna H, Weigel BJ, Taylor PA, Blazar BR. In vitro tumor-pulsed or in vivo Flt3 ligand-generated dendritic cells provide protection against acute myelogenous leukemia in nontransplanted or syngeneic bone marrow-transplanted mice. Blood 2001; 97:1474-82. [PMID: 11222396 DOI: 10.1182/blood.v97.5.1474] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To determine whether immune stimulation could reduce acute myelogenous leukemia (AML) lethality, dendritic cells (DCs) were pulsed with AML antigens and used as vaccines or generated in vivo by Flt3 ligand (Flt3L), a potent stimulator of DC and natural killer (NK) cell generation. Mice were then challenged with AML cells. The total number of splenic anti-AML cytotoxic T-lymphocyte precursors (CTLPs) present at the time of challenge was increased 1.9-fold and 16.4-fold by Flt3L or DC tumor vaccines, respectively. As compared with the 0% survival of controls, 63% or more of recipients of pulsed DCs or Flt3L survived long term. Mice given AML cells prior to DC vaccines or Flt3L had only a slight survival advantage versus non-treated controls. NK cells or NK cells and T cells were found to be involved in the antitumor responses of Flt3L or DCs, respectively. DC vaccines lead to long-term memory responses but Flt3L does not. Syngeneic bone marrow transplantation (BMT) recipients were analyzed beginning 2 months post-BMT. In contrast to the uniform lethality in BMT controls given AML cells, recipients of either Flt3L or DC vaccines had a significant increase in survival. The total number of splenic anti-AML CTLPs at the time of AML challenge in BMT controls was 40% of concurrently analyzed non-BMT controls. Flt3L or DC vaccines increased the total anti-AML CTLPs 1.4-fold and 6.8-fold, respectively. Neither approach was successful when initiated after AML challenge. It was concluded that DC vaccines and Flt3L administration can enhance an AML response in non-transplanted or syngeneic BMT mice but only when initiated prior to AML progression.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/pharmacology
- Animals
- Antibody Formation/radiation effects
- Bone Marrow Transplantation
- CD8-Positive T-Lymphocytes/immunology
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/transplantation
- Hematopoiesis/drug effects
- Immunotherapy, Adoptive
- Killer Cells, Natural/immunology
- Leukemia, Myeloid, Acute/prevention & control
- Leukemia, Myeloid, Acute/therapy
- Membrane Proteins/administration & dosage
- Membrane Proteins/pharmacology
- Mice
- Mice, Inbred C57BL
- Neoplasm Transplantation
- Transplantation, Isogeneic
- Tumor Cells, Cultured/transplantation
Collapse
Affiliation(s)
- A B Pawlowska
- University of Minnesota Cancer Center and Department of Pediatrics, Division of Bone Marrow Transplantation, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | |
Collapse
|
45
|
Abstract
Apoptotic death of CD8+ T cells can be induced by a population of inhibitory myeloid cells that are double positive for the CD11b and Gr-1 markers. These cells are responsible for the immunosuppression observed in pathologies as dissimilar as tumor growth and overwhelming infections, or after immunization with viruses. The appearance of a CD11b+/Gr-1+ population of inhibitory macrophages (iMacs) could be attributed to high levels of granulocyte-macrophage colony-stimulating factor (GM-CSF) in vivo. Deletion of iMacs in vitro or in vivo reversed the depression of CD8+ T-cell function. We isolated iMacs from the spleens of immunocompromised mice and found that these cells were positive for CD31, ER-MP20 (Ly-6C), and ER-MP58, markers characteristic of granulocyte/monocyte precursors. Importantly, although iMacs retained their inhibitory properties when cultured in vitro in standard medium, suppressive functions could be modulated by cytokine exposure. Whereas culture with the cytokine interleukin 4 (IL-4) increasediMac inhibitory activity, these cells could be differentiated into a nonadherent population of fully mature and highly activated dendritic cells when cultured in the presence of IL-4and GM-CSF. A common CD31+/CD11b+/Gr-1+ progenitor can thus give rise to cells capable of either activating or inhibiting the function of CD8+ T lymphocytes, depending on the cytokinemilieu that prevails during antigen-presenting cell maturation.
Collapse
|
46
|
Identification of a CD11b+/Gr-1+/CD31+ myeloid progenitor capable of activating or suppressing CD8+T cells. Blood 2000. [DOI: 10.1182/blood.v96.12.3838] [Citation(s) in RCA: 362] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Apoptotic death of CD8+ T cells can be induced by a population of inhibitory myeloid cells that are double positive for the CD11b and Gr-1 markers. These cells are responsible for the immunosuppression observed in pathologies as dissimilar as tumor growth and overwhelming infections, or after immunization with viruses. The appearance of a CD11b+/Gr-1+ population of inhibitory macrophages (iMacs) could be attributed to high levels of granulocyte-macrophage colony-stimulating factor (GM-CSF) in vivo. Deletion of iMacs in vitro or in vivo reversed the depression of CD8+ T-cell function. We isolated iMacs from the spleens of immunocompromised mice and found that these cells were positive for CD31, ER-MP20 (Ly-6C), and ER-MP58, markers characteristic of granulocyte/monocyte precursors. Importantly, although iMacs retained their inhibitory properties when cultured in vitro in standard medium, suppressive functions could be modulated by cytokine exposure. Whereas culture with the cytokine interleukin 4 (IL-4) increasediMac inhibitory activity, these cells could be differentiated into a nonadherent population of fully mature and highly activated dendritic cells when cultured in the presence of IL-4and GM-CSF. A common CD31+/CD11b+/Gr-1+ progenitor can thus give rise to cells capable of either activating or inhibiting the function of CD8+ T lymphocytes, depending on the cytokinemilieu that prevails during antigen-presenting cell maturation.
Collapse
|
47
|
Abstract
The survival of patients with cancer has improved steadily but incrementally over the last century, with the advent of effective anticancer treatments such as chemotherapy and radiotherapy. However, the majority of patients with metastatic disease will not be cured by these measures and will eventually die of their disease. New and more effective methods of treating these patients are required urgently. The immune system is a potent force for rejecting transplanted organs or microbial pathogens, but effective spontaneous immunologically induced cancer remissions are very rare. In recent years, much has been discovered about the mechanisms by which the immune system recognizes and responds to cancers. The specific antigens involved have now been defined in many cases. Improved adjuvants are available. Means by which cancer cells overcome immunological attack can be exploited and overcome. Most importantly, the immunological control mechanisms responsible for initiating and maintaining an effective immune response are now much better understood. It is now possible to manipulate immunological effector cells or antigen-presenting cells ex vivo in order to induce an effective antitumour response. At the same time, it is possible to recruit other aspects of the immune system, both specific (e.g. antibody responses) and innate (natural killer cells and granulocytes).
Collapse
Affiliation(s)
- I D Davis
- Ludwig Institute for Cancer Research, Austin Repat Cancer Centre, Heidelberg, Victoria, Australia.
| |
Collapse
|
48
|
Driessen C, Bryant RA, Lennon-Duménil AM, Villadangos JA, Bryant PW, Shi GP, Chapman HA, Ploegh HL. Cathepsin S controls the trafficking and maturation of MHC class II molecules in dendritic cells. J Cell Biol 1999; 147:775-90. [PMID: 10562280 PMCID: PMC2156161 DOI: 10.1083/jcb.147.4.775] [Citation(s) in RCA: 191] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/1999] [Accepted: 10/01/1999] [Indexed: 12/30/2022] Open
Abstract
Before a class II molecule can be loaded with antigenic material and reach the surface to engage CD4+ T cells, its chaperone, the class II-associated invariant chain (Ii), is degraded in a stepwise fashion by proteases in endocytic compartments. We have dissected the role of cathepsin S (CatS) in the trafficking and maturation of class II molecules by combining the use of dendritic cells (DC) from CatS(-/-) mice with a new active site-directed probe for direct visualization of active CatS. Our data demonstrate that CatS is active along the entire endocytic route, and that cleavage of the lysosomal sorting signal of Ii by CatS can occur there in mature DC. Genetic disruption of CatS dramatically reduces the flow of class II molecules to the cell surface. In CatS(-/-) DC, the bulk of major histocompatibility complex (MHC) class II molecules is retained in late endocytic compartments, although paradoxically, surface expression of class II is largely unaffected. The greatly diminished but continuous flow of class II molecules to the cell surface, in conjunction with their long half-life, can account for the latter observation. We conclude that in DC, CatS is a major determinant in the regulation of intracellular trafficking of MHC class II molecules.
Collapse
Affiliation(s)
- Christoph Driessen
- Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115
| | - Rebecca A.R. Bryant
- Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115
| | | | - José A. Villadangos
- Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115
| | - Paula Wolf Bryant
- Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Harold A. Chapman
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Hidde L. Ploegh
- Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|