1
|
Diversity of dendritic cells generated from umbilical cord or adult peripheral blood precursors. Cent Eur J Immunol 2018; 43:306-313. [PMID: 30588175 PMCID: PMC6305608 DOI: 10.5114/ceji.2018.80050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 04/24/2017] [Indexed: 02/01/2023] Open
Abstract
Following the discovery of methods to generate large numbers of specific dendritic cells (DCs) ex vivo, the possibility of exploiting these cells in immunotherapeutic strategies will become a reality. It seems to be rationally to analyse the influence of the precursor source for further features and applications. For the needs of the given project DCs were derived from precursors derived from adult peripheral blood (APB) and umbilical cord blood (UCB). During some expansions of UCB CD34+ cells were separated giving non-adherent DCs (NA-DCs) or adherent DCs (A-DCs), whereas DCs derived from UCB precursors without separation gave rise to All-DCs. DC subpopulations were stimulated by lipopolysaccharides (LPS) or interferon-γ (IFN-γ), and afterwards the morphology, phenotype, and stimulatory properties were analysed. Our findings demonstrated that DCs generated from APB and UCB precursors were not equivalent and exhibited opposite features when expanded in comparable conditions. Additionally, all three subpopulations of UCB-derived DCs presented functional dissimilarities. Based on our results we concluded that the precursor source and the composition of media must be considered as crucial to the success of potential therapeutic application.
Collapse
|
2
|
Kim KH, Kim SH, Jung HH, Moon JH, Jeong SU, Yu K, Lee CK. Thapsigargin Increases IL-2 Production in T Cells at Nanomolar Concentrations. Immune Netw 2018; 18:e26. [PMID: 30181914 PMCID: PMC6117510 DOI: 10.4110/in.2018.18.e26] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/27/2018] [Accepted: 07/30/2018] [Indexed: 12/02/2022] Open
Abstract
Thapsigargin (TGN) is a potent and selective inhibitor of sarco-endoplasmic Ca2+-ATPase, leading to rapid elevation of cytoplasmic Ca2+ concentration. Previous reports have shown that TGN increases the production of various cytokines from macrophages and dendritic cells. Here, we examine the effects of TGN on murine T cells. Nanomolar concentrations of TGN are a significant inducer of IL-2 production with full activity at 50 nM. Micromolar concentrations of TGN, however, are inhibitory to IL-2 production and T cell proliferation. The IL-2 production-inducing activity of TGN is much more prominent when T cells are primed with concanavalin A or anti-CD3 mAb, and is due to the increase of cytoplasmic Ca2+ concentration. TGN at 50 nM does not affect interferon-gamma or IL-4 production from T cells. Thus, the present study shows that low nanomolar concentrations of TGN could be useful in potentiating IL-2 production from antigen-primed T cells.
Collapse
Affiliation(s)
- Ki-Hyang Kim
- College of Pharmacy, Chungbuk National University, Cheongju 28644, Korea
| | - Sang-Hyun Kim
- College of Pharmacy, Chungbuk National University, Cheongju 28644, Korea
| | - Ho-Hyun Jung
- College of Pharmacy, Chungbuk National University, Cheongju 28644, Korea
| | - Jun-Hyeok Moon
- College of Pharmacy, Chungbuk National University, Cheongju 28644, Korea
| | - Seong-Un Jeong
- College of Pharmacy, Chungbuk National University, Cheongju 28644, Korea
| | - Kyeongae Yu
- College of Pharmacy, Chungbuk National University, Cheongju 28644, Korea
| | - Chong-Kil Lee
- College of Pharmacy, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
3
|
Vázquez MB, Sureda M, Rebollo J. Células dendríticas I: aspectos básicos de su biología y funciones. ACTA ACUST UNITED AC 2012. [DOI: 10.1016/j.inmuno.2011.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
4
|
Tuyaerts S, Aerts JL, Corthals J, Neyns B, Heirman C, Breckpot K, Thielemans K, Bonehill A. Current approaches in dendritic cell generation and future implications for cancer immunotherapy. Cancer Immunol Immunother 2007; 56:1513-37. [PMID: 17503040 PMCID: PMC11030932 DOI: 10.1007/s00262-007-0334-z] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2007] [Accepted: 04/17/2007] [Indexed: 02/06/2023]
Abstract
The discovery of tumor-associated antigens, which are either selectively or preferentially expressed by tumors, together with an improved insight in dendritic cell biology illustrating their key function in the immune system, have provided a rationale to initiate dendritic cell-based cancer immunotherapy trials. Nevertheless, dendritic cell vaccination is in an early stage, as methods for preparing tumor antigen presenting dendritic cells and improving their immunostimulatory function are continuously being optimized. In addition, recent improvements in immunomonitoring have emphasized the need for careful design of this part of the trials. Still, valuable proofs-of-principle have been obtained, which favor the use of dendritic cells in subsequent, more standardized clinical trials. Here, we review the recent developments in clinical DC generation, antigen loading methods and immunomonitoring approaches for DC-based trials.
Collapse
Affiliation(s)
- Sandra Tuyaerts
- Laboratory of Molecular and Cellular Therapy, Department of Physiology and Immunology, Medical School of the Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090 Brussels, Belgium
| | - Joeri L. Aerts
- Laboratory of Molecular and Cellular Therapy, Department of Physiology and Immunology, Medical School of the Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090 Brussels, Belgium
| | - Jurgen Corthals
- Laboratory of Molecular and Cellular Therapy, Department of Physiology and Immunology, Medical School of the Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090 Brussels, Belgium
| | - Bart Neyns
- Medical Oncology, Oncology Center, University Hospital Brussels, Free University Brussels, Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Carlo Heirman
- Laboratory of Molecular and Cellular Therapy, Department of Physiology and Immunology, Medical School of the Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090 Brussels, Belgium
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy, Department of Physiology and Immunology, Medical School of the Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090 Brussels, Belgium
| | - Kris Thielemans
- Laboratory of Molecular and Cellular Therapy, Department of Physiology and Immunology, Medical School of the Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090 Brussels, Belgium
| | - Aude Bonehill
- Laboratory of Molecular and Cellular Therapy, Department of Physiology and Immunology, Medical School of the Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090 Brussels, Belgium
| |
Collapse
|
5
|
Abstract
Dendritic cells (DCs) play a crucial role in the induction of antigen-specific T-cell responses, and therefore their use for the active immunotherapy of malignancies has been studied with considerable interest. More than a decade has passed since the publication of the first clinical data of DC-based vaccines, and through this and subsequent studies, a number of important developmental insights have been gleaned. These include the ideal source and type of DCs, the discovery of novel antigens and methods of loading DCs, the role of DC maturation, and the most efficient route of immunization. The generation of immune responses against tumor antigens after DC immunization has been demonstrated, and favorable clinical responses have been reported in some patients; however, it is difficult to pool the results as a whole, and thus the body of data remains inconclusive, in part because of varying DC preparation and vaccination protocols, the use of different forms of antigens, and, most importantly, a lack of rigorous criteria for defining clinical responses. As such, the standardization of clinical and immunologic criteria utilized, as well as DC preparations employed, will allow for the comparison of results across multiple clinical studies and is required in order for future trials to measure the true value and role of this treatment modality. In addition, issues regarding the optimal dose and clinical setting for the application of DC vaccines remain to be resolved, and recent clinical studies have been designed to begin to address these questions.
Collapse
Affiliation(s)
- Takuya Osada
- Department of Surgery, Program in Molecular Therapeutics, Comprehensive Cancer Center, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | |
Collapse
|
6
|
Smith JW, Gamelli RL, Jones SB, Shankar R. Immunologic responses to critical injury and sepsis. J Intensive Care Med 2006; 21:160-72. [PMID: 16672638 DOI: 10.1177/0885066605284330] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Almost 2 million patients are admitted to hospitals in the United States each year for treatment of traumatic injuries, and these patients are at increased risk of late infections and complications of systemic inflammation as a result of injury. Host response to injury involves a general activation of multiple systems in defending the organism from hemorrhagic or infectious death. Clinicians have the capability to support the critically injured through their traumatic insult with surgery and improved critical care, but the inflammatory response generated by such injuries creates new challenges in the management of these patients. It has long been known that local tissue injury induces systemic changes in the traumatized patient that are often maladaptive. This article reviews the effects of injury on the function of immune system cells and highlights some of the clinical sequelae of this deranged inflammatory-immune interaction.
Collapse
Affiliation(s)
- Jason W Smith
- Department of Surgery and Burn & Shock Trauma Institute, Loyola University Medical Center, Maywood, IL 60153, USA
| | | | | | | |
Collapse
|
7
|
Moldenhauer A, Moore MAS, Schmidt K, Kiesewetter H, Salama A. Differences in the transmigration of different dendritic cells. Exp Hematol 2006; 34:745-52. [PMID: 16728279 DOI: 10.1016/j.exphem.2006.02.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2005] [Revised: 02/01/2006] [Accepted: 02/08/2006] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Although several methods for the generation of dendritic cells (DCs) exist, little is known about the transmigration capacities of the cells developed. Their ability to migrate to the adjacent lymphatic system is relevant since their efficacy does also rely on their potential to interact with lymphocytes. METHODS We studied the transmigration of DCs derived from hematopoietic progenitor cells (HPC), from peripheral blood monocytes, and from leukemic cells. DCs from monocytes and leukemic cells could be generated within 1 week, whereas DCs from HPC needed 2 weeks for maturation. RESULTS While DCs from all sources showed similar morphologic features and allostimulatory capacities, their transmigration capacities varied: HPC-derived DCs showed the highest migratory response to macrophage inflammatory protein (MIP)-3alpha and beta. Monocyte-derived DCs were equally attracted to MIP-3beta and stroma-derived factor (SDF)-1alpha. Only few leukemic DCs migrated in response to SDF-1. Other chemoattractants tested included MIP-1alpha and RANTES. Replacement of fetal bovine by human serum did not change the DC's overall migratory capacities. It did, however, influence the responsiveness to certain chemokines. CONCLUSION Although DCs from all three sources are immunocompetent antigen-presenting cells, our findings suggest that HPC and monocyte-derived DCs can be administered subcutaneously and intravenously, but that leukemic DCs should be injected into the lymph node.
Collapse
Affiliation(s)
- Anja Moldenhauer
- Institute for Transfusion Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | | | | | | | | |
Collapse
|
8
|
Liau LM, Prins RM, Kiertscher SM, Odesa SK, Kremen TJ, Giovannone AJ, Lin JW, Chute DJ, Mischel PS, Cloughesy TF, Roth MD. Dendritic cell vaccination in glioblastoma patients induces systemic and intracranial T-cell responses modulated by the local central nervous system tumor microenvironment. Clin Cancer Res 2006; 11:5515-25. [PMID: 16061868 DOI: 10.1158/1078-0432.ccr-05-0464] [Citation(s) in RCA: 401] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We previously reported that autologous dendritic cells pulsed with acid-eluted tumor peptides can stimulate T cell-mediated antitumor immune responses against brain tumors in animal models. As a next step in vaccine development, a phase I clinical trial was established to evaluate this strategy for its feasibility, safety, and induction of systemic and intracranial T-cell responses in patients with glioblastoma multiforme. EXPERIMENTAL DESIGN Twelve patients were enrolled into a multicohort dose-escalation study and treated with 1, 5, or 10 million autologous dendritic cells pulsed with constant amounts (100 mug per injection) of acid-eluted autologous tumor peptides. All patients had histologically proven glioblastoma multiforme. Three biweekly intradermal vaccinations were given; and patients were monitored for adverse events, survival, and immune responses. The follow-up period for this trial was almost 5 years. RESULTS Dendritic cell vaccinations were not associated with any evidence of dose-limiting toxicity or serious adverse effects. One patient had an objective clinical response documented by magnetic resonance imaging. Six patients developed measurable systemic antitumor CTL responses. However, the induction of systemic effector cells did not necessarily translate into objective clinical responses or increased survival, particularly for patients with actively progressing tumors and/or those with tumors expressing high levels of transforming growth factor beta(2) (TGF-beta(2)). Increased intratumoral infiltration by cytotoxic T cells was detected in four of eight patients who underwent reoperation after vaccination. The magnitude of the T-cell infiltration was inversely correlated with TGF-beta(2) expression within the tumors and positively correlated with clinical survival (P = 0.047). CONCLUSIONS Together, our results suggest that the absence of bulky, actively progressing tumor, coupled with low TGF-beta(2) expression, may identify a subgroup of glioma patients to target as potential responders in future clinical investigations of dendritic cell-based vaccines.
Collapse
Affiliation(s)
- Linda M Liau
- Division of Neurosurgery, Department of Surgery, The Brain Research Institute, David Geffen School of Medicine at University of California at Los Angeles, University of California Los Angeles, Los Angeles, California 90095, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Isotani M, Katsuma K, Tamura K, Yamada M, Yagihara H, Azakami D, Ono K, Washizu T, Bonkobara M. Efficient Generation of Canine Bone Marrow-Derived Dendritic Cells. J Vet Med Sci 2006; 68:809-14. [PMID: 16953080 DOI: 10.1292/jvms.68.809] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Because of their unsurpassed potency in presenting antigens to naive T cells, dendritic cells are considered to be an important candidate in the development of immunotherapeutic strategies. Despite the high potential of dendritic cell-based immunotherapy, as a so-called dendritic cell vaccination, few clinical approaches using dendritic cell vaccination have been performed in the dog because of very limited information regarding the generation of canine dendritic cells and their functional properties. We therefore established a protocol for the efficient generation of dendritic cells from canine bone marrow cells using recombinant feline granulocyte-macrophage colony-stimulating factor and canine interleukin-4. Dendritic cells were generated efficiently: a yield of 1-9 x 10(6) cells per approximately 0.5 ml of canine bone marrow aspiration was achieved. These dendritic cells showed features shared with mouse and human dendritic cells: dendrite morphology, expression of surface markers MHC class II and CD11c, and up-regulation of molecules related to antigen presentation (MHC class II, B7-1, and B7-2) by activation with lipopolysaccharide. Moreover, the dendritic cells demonstrated phagocytic activity, processing activity of pinocytosed proteins, and activation of allogeneic T cells far more potent than that by macrophages. Our findings suggest that the bone marrow-derived dendritic cells are functional for the capturing and processing of antigens and the initiation of T cell responses.
Collapse
Affiliation(s)
- Mayu Isotani
- Department of Veterinary Clinical Pathology, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Syme R, Bajwa R, Robertson L, Stewart D, Glück S. Comparison of CD34 and monocyte-derived dendritic cells from mobilized peripheral blood from cancer patients. Stem Cells 2005; 23:74-81. [PMID: 15625124 DOI: 10.1634/stemcells.2004-0070] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Dendritic cells (DCs) are potent antigen-presenting cells that are integral to the initiation of T-cell immunity. Two cell types can be used as a source for generating DCs: monocytes and CD34(+) stem cells. Despite many investigations characterizing DCs, none have performed a direct paired comparison of monocyte and stem cell-derived DCs. Therefore, it is unclear whether one cell source has particular advantages over the other, or whether inherent differences exist between the two populations. We undertook the following study to determine if there were any differences in DCs generated from monocytes or CD34(+) cells from mobilized peripheral blood. DCs were generated by culturing the adherent cells (monocytes) in interleukin-4 and GM-CSF for 7 days, or by culturing nonadherent cells (CD34(+)) in the presence of GM-CSF and tumor necrosis factor alpha for 14 days. The resulting DCs were compared morphologically, phenotypically, functionally, and by yield. We could generate morphologically and phenotypically similar DCs. Differences were encountered when expression levels of some cell surface markers were examined (CD86, HLA-DR). There was no difference in how the DCs performed in a mixed lymphocyte reaction (p = .3). Further, no statistical difference was discovered when we examined cellular (DC) yield (p = .1); however, there was a significant difference when yield was normalized to the starting number of monocytes or CD34(+) cells (p = .016). Together, these data demonstrate that differences do exist between monocyte-derived DCs and CD34-derived DCs from the same cellular product (apheresis) from the same individual.
Collapse
Affiliation(s)
- Rachel Syme
- Clinical Research Program, Department of Oncology, Tom Baker Cancer Centre, 1331 29th Street NW, Calgary, AB, T2N 4N2, Canada.
| | | | | | | | | |
Collapse
|
11
|
Schütt P, Buttkereit U, Brandhorst D, Lindemann M, Schmiedl S, Grosse-Wilde H, Seeber S, Nowrousian MR, Opalka B, Moritz T. In vitro dendritic cell generation and lymphocyte subsets in myeloma patients: influence of thalidomide and high-dose chemotherapy treatment. Cancer Immunol Immunother 2005; 54:506-12. [PMID: 15750834 PMCID: PMC11032805 DOI: 10.1007/s00262-004-0633-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2004] [Accepted: 09/30/2004] [Indexed: 10/26/2022]
Abstract
While vaccination with antigen-pulsed dendritic cells (DCs) represents a promising therapeutic strategy in multiple myeloma (MM), clinical benefit, so far, has been limited to individual patients. To identify potential problems with this approach, we have analyzed the influence of treatment parameters, in particular high-dose chemotherapy (HD-CTX) and thalidomide, on in vitro DC generation and peripheral blood lymphocyte subsets in MM patients. From a total of 25 MM patients, including 14 patients on thalidomide treatment and 11 after HD-CTX, in vitro DC generation from peripheral blood monocytes under serum-free condition was investigated. In addition, peripheral blood lymphocyte subsets were assessed in 17 patients including 10 patients on thalidomide treatment and 9 patients after HD-CTX. Efficient in vitro generation of DCs (median 7.1x10(6)/100 ml peripheral blood; range 0.1-42.5x10(6)/100 ml peripheral blood) expressing DC-typical surface markers was observed in 23 MM patients (92%), although reduced expression of CD1a, CD40, CD83, and HLA-DR was observed in patients treated with thalidomide. With respect to lymphocyte subsets, MM patients showed significantly (p<0.05) reduced B and CD4+ lymphocytes in the peripheral blood. This effect was most prominent within 6 months of HD-CTX and in patients receiving thalidomide (usually in combination with CTX). CD8+ lymphocytes were significantly increased in MM patients. Thus, despite the well-known deficiencies in their immune system, adequate numbers of DCs can be generated in most myeloma patients. In patients treated with thalidomide, however, it remains to be seen whether the reduced expression of co-stimulatory molecules has functional relevance.
Collapse
Affiliation(s)
- Philipp Schütt
- Department of Internal Medicine (Cancer Research), West German Cancer Center, University of Duisburg-Essen Medical School, Hufelandstrasse 55, 45122 Essen, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Tokunaga N, Murakami T, Endo Y, Nishizaki M, Kagawa S, Tanaka N, Fujiwara T. Human Monocyte-Derived Dendritic Cells Pulsed with Wild-Type p53 Protein Efficiently Induce CTLs against p53 Overexpressing Human Cancer Cells. Clin Cancer Res 2005. [DOI: 10.1158/1078-0432.1312.11.3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Purpose: Dendritic cells are the most potent antigen-presenting cells for initiating cellular immune responses. Dendritic cells are attractive immunoregulatory cells for cancer immunotherapy, and their efficacy has been investigated in clinical trials. The tumor suppressor gene p53 is pivotal in the regulation of apoptosis, and p53-based immunization is an attractive approach to cancer immunotherapy because of the accumulation of p53 protein in malignant but not in normal cells. It has been shown that dendritic cells transduced with an adenoviral wild-type p53 (wt-p53) construct mediate the antitumor immune responses against p53-overexpressing tumor cells. We examined whether monocyte-derived human dendritic cells pulsed with the purified full-length wt-p53 protein were also capable of inducing the specific antitumor responses against p53-overexpressing tumors in vitro.
Experimental Design: Immature dendritic cells generated in the presence of interleukin-4 and granulocyte/macrophage colony-stimulating factor from monocytes of HLA-A2- or HLA-A24-positive healthy individuals were pulsed with the purified p53 protein. Uptake of p53 protein by human dendritic cells was assessed by Western blotting and immunohistochemical staining using anti-p53 antibody. Induction of p53-specific CTL response was also evaluated by the cytotoxic assay against p53-overexpressing human tumor cells.
Results: Both Western blot and immunohistochemical analysis showed the accumulation of p53 protein in human immature dendritic cells. T cells obtained from HLA-A2- or HLA-A24-positive healthy donors were stimulated twice with p53 protein-pulsed dendritic cells and then applied to the cytotoxicity assay against p53-overexpressing target cells. The CTL activity was specific for p53-overexpressing tumor cells and MHC class I restricted. Moreover, the CTL activity generated by p53 protein-pulsed dendritic cells was nearly identical with that induced by adenoviral wt-p53-transduced dendritic cells.
Conclusions: Our results indicate that monocyte-derived human dendritic cells pulsed with the wt-p53 protein could induce the specific antitumor effect against p53-overexpressing tumors and that this in vitro model offers a new and more simple approach to the development of p53-based immunotherapy.
Collapse
Affiliation(s)
- Naoyuki Tokunaga
- 1Division of Surgical Oncology, Department of Surgery, Okayama University Graduate School of Medicine and Dentistry, and
| | - Takayoshi Murakami
- 1Division of Surgical Oncology, Department of Surgery, Okayama University Graduate School of Medicine and Dentistry, and
| | - Yoshikatsu Endo
- 1Division of Surgical Oncology, Department of Surgery, Okayama University Graduate School of Medicine and Dentistry, and
| | - Masahiko Nishizaki
- 1Division of Surgical Oncology, Department of Surgery, Okayama University Graduate School of Medicine and Dentistry, and
| | - Shunsuke Kagawa
- 2Center for Gene and Cell Therapy, Okayama University Hospital, Okayama, Japan
| | - Noriaki Tanaka
- 1Division of Surgical Oncology, Department of Surgery, Okayama University Graduate School of Medicine and Dentistry, and
| | - Toshiyoshi Fujiwara
- 1Division of Surgical Oncology, Department of Surgery, Okayama University Graduate School of Medicine and Dentistry, and
- 2Center for Gene and Cell Therapy, Okayama University Hospital, Okayama, Japan
| |
Collapse
|
13
|
Misawa E, Sakurai T, Yamada M, Tamura Y, Motoyoshi K. Administration of macrophage colony-stimulating factor mobilized both CD11b+CD11c+ cells and NK1.1+ cells into peripheral blood. Int Immunopharmacol 2004; 4:791-803. [PMID: 15135320 DOI: 10.1016/j.intimp.2004.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2004] [Revised: 01/22/2004] [Accepted: 03/04/2004] [Indexed: 11/23/2022]
Abstract
We attempted the phenotypic characterization of peripheral blood (PB) cells after daily administration of macrophage colony-stimulating factor (M-CSF) in mice. The number of CD11b+ cells was increased by M-CSF treatment (2- and 5-day injections). Notably, CD11bbrightCD11cdim, CD11b+CD11c+ and CD11b+CD80+ cells were significantly increased by 2-day treatment of M-CSF. On the other hand, the number of NK1.1+ cells was not changed by the 2-day treatment, but it was significantly increased by the 5-day treatment. However, the numbers of CD3+ and NK1.1+CD3+ cells were not changed by M-CSF treatment. Then, mononuclear cells (MNCs) were separated from the PB of mice treated with saline or M-CSF, and they were incubated with GM-CSF + IL-4 or IL-2. Compared with the saline-treated one (S-MNCs), the MNCs of M-CSF-treated mice (M-MNCs) showed strong proliferation by the GM-CSF + IL-4 stimulation. The MNCs could stimulate proliferation of allo-T cells in the mixed lymphocyte reaction (MLR), especially the M-MNCs showed strong reaction. On the other hand, the stimulation by IL-2 induced strong cell growth of MNCs. And M-CSF treatment enhanced this response. Furthermore, the M-MNCs (stimulated by IL-2 in vitro) exhibited greater cytotoxicity against Yac-1 cells than the S-MNCs. In conclusion, we found that administration of M-CSF mobilized CD11b+, CD11b+CD11c+, CD11b+CD80+, and NK1.1+cells into PB. And the injection of M-CSF facilitates the generation of dendritic and natural killer cells from PB cells in vitro. These results suggest that the mobilized cells may provide for application of immunotherapy.
Collapse
Affiliation(s)
- Eriko Misawa
- Biochemical Research Laboratory, Morinaga Milk Industry Co. Ltd., Higashihara 5-1-83, Zama, Kanagawa 228-8583, Japan.
| | | | | | | | | |
Collapse
|
14
|
Mosca PJ, Clay TM, Kim Lyerly H, Morse MA. Current status of dendritic cell immunotherapy of malignancies. Int Rev Immunol 2003; 22:255-81. [PMID: 12745642 DOI: 10.1080/08830180305223] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Because dendritic cells (DC) are central to the induction of antigen-specific T cell responses, their use for the active immunotherapy of malignancies has been of considerable interest. Since clinical trials with DC-based vaccines have been initiated, a number of important developmental issues have become apparent. These include the ideal source and type of DC, the form of antigen and method of loading DC, whether to induce maturation, the route and timing of immunization, and the optimal clinical scenario. Clinical responses such as stability of disease and tumor regressions have been reported in some patients, particularly with melanoma, myeloma, and prostate cancer.
Collapse
Affiliation(s)
- Paul J Mosca
- Departments of Surgery and Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | | |
Collapse
|
15
|
Curry JL, Qin JZ, Bonish B, Carrick R, Bacon P, Panella J, Robinson J, Nickoloff BJ. Innate immune-related receptors in normal and psoriatic skin. Arch Pathol Lab Med 2003; 127:178-86. [PMID: 12562231 DOI: 10.5858/2003-127-178-iirrin] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
CONTEXT A precise role for the innate immune system in psoriasis remains to be determined. Surface receptors, including Toll-like receptors (TLRs) that recognize bacterial ligands and CD91, which recognizes heat shock proteins (HSPs), are implicated in both innate and adaptive immunity. OBJECTIVE Since skin is exposed to various exogenous stimuli, which can provoke or exacerbate psoriasis, we characterized expression and function of TLRs, CD91, and HSPs in normal and psoriatic skin. DESIGN A variety of skin-derived cells and blood-derived cells were analyzed both in vivo and in vitro; samples were obtained from 24 different individuals for innate immune-related receptor expression and function. By comparing and contrasting individuals with healthy skin and psoriatic patients, several specific differences were identified. RESULTS Immunohistochemistry-based expression profiling revealed TLR1 expression in epidermal dendritic cells (DCs) and dermal dendritic cells (DDCs) in normal skin, as well as in pre-psoriatic skin and psoriatic plaques, with enhanced basal layer keratinocyte (KC) expression in pre-psoriatic and psoriatic plaques compared with normal skin; TLR2 expression primarily by DDCs; and TLR4 expression by epidermal DCs and DDCs, with mid-epidermal-layer KCs displaying cell surface staining. No TLR9 or CD14 was detected on DCs or KCs, although psoriatic plaques contained CD14-positive macrophages. Analysis of psoriatic epidermis revealed HSPs 27, 60, and 70. Keratinocytes were CD91 negative, but CD91 was expressed by fibroblasts and DDCs in normal and pre-psoriatic skin, with prominent accumulation of CD91-positive DDCs in psoriatic plaques. Cultured KCs revealed no surface expression of TLR2, TLR4, TLR9, or CD91. Exposure of fibroblasts, but not KCs, to lipopolysaccharide or HSPs triggered nuclear factor (NF)-kappaB activation. Heat shock proteins did induce maturation of blood-derived DCs accompanied by increased interleukin-12 production and enhanced antigen-presenting function. CONCLUSIONS These data demonstrate distinctive patterns of innate immune-related receptors by specific subsets of cells in normal and psoriatic skin, suggesting functional roles for HSPs and DCs in psoriasis.
Collapse
Affiliation(s)
- Jonathan L Curry
- Department of Pathology, Skin Cancer Research Laboratories, Loyola University Cancer Center, Maywood, IL 60153, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Joshi SS, Vu UE, Lovgren TR, Lorkovic M, Patel W, Todd GL, Kuszynski C, Joshi BJ, Dave HP. Comparison of phenotypic and functional dendritic cells derived from human umbilical cord blood and peripheral blood mononuclear cells. JOURNAL OF HEMATOTHERAPY & STEM CELL RESEARCH 2002; 11:337-47. [PMID: 11983105 DOI: 10.1089/152581602753658529] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Dendritic cells (DC) are important accessory cells that are capable of initiating an immune response. Generation of functional DC has potential clinical use in treating diseases such as cancer. In this report, we have demonstrated the generation of functional DC from mononuclear cells isolated from human umbilical cord blood cells (UCBC) and peripheral blood cells (PBC) using a defined medium Prime Complete Growth Medium (PCGM) (GenePrime LLC, Gaithersburg, MD). DC generated using PCGM showed the typical phenotype of DC as determined by flow cytometry and electron microscopy. Further analysis of the DC using confocal microscopy showed localization of the antigen and major histocompatibility complex (MHC) molecules in the cytoplasm 3-5 days following tumor antigen loading into DC. Subsequently, the tumor antigen-MHC complex was localized on the surface of DC. DC generated from UCBC or PBC also increased (p < 0.001) the allogeneic mixed lymphocyte reaction, confirming their immune accessory functions compared to a control mixed lymphocyte reaction (MLR) without DC added. Interestingly, DC generated using PCGM medium also significantly enhanced the hematopoietic colony (CFU-C)-forming ability. Furthermore, addition of 5% DC derived from cord blood loaded with tumor antigen also significantly (p < 0.001) increased peripheral and cord blood-derived antigen-specific cytotoxic T lymphocyte (CTL)-mediated killing of human leukemic cells (K562) and breast cancer cells (MDA-231). Thus, these results show that functional DC generated from cord blood using a defined medium are a useful source of accessory cells for augmenting CTL-mediated cytotoxicity and have potential use in cellular therapy for human leukemia and breast cancer.
Collapse
Affiliation(s)
- Shantaram S Joshi
- Department of Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198-6395, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|