1
|
Liu G, Fan X, Cai Y, Fu Z, Gao F, Dong J, Li K, Cai J. Efficacy of dendritic cell-based immunotherapy produced from cord blood in vitro and in a humanized NSG mouse cancer model. Immunotherapy 2019; 11:599-616. [PMID: 30943862 DOI: 10.2217/imt-2018-0103] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 02/18/2019] [Indexed: 02/08/2023] Open
Abstract
AIM To produce dendritic cells (DCs) from CD34+ stem cells from cord blood and explore their prophylactic and curative effect against tumors by vaccinating humanized NSG mice. MATERIALS & METHODS Separated CD34+ stem cells from cord blood were cultured for 30 days, and the resultant DCs (CD34-DCs) were collected. The basic function of the CD34-DCs and the cytotoxicity of CD34-cytotoxic-T lymphocytes (CTLs) were tested in vitro, and tumor inhibition in a humanized NSG mouse tumor model was observed. RESULTS The number of CD34-DCs reached approximately 9 log. These cells performed functions similar to those of DCs derived from monocytes from peripheral blood (PBMC-DCs). The CTLs of the CD34-DCs (CD34-CTLs) presented a better antitumor effect in vitro. The obvious prophylactic and therapeutic antitumor effects of the CD34-DC vaccine were observed in the humanized NSG mouse models. CONCLUSION CD34-DCs from cord blood were sufficient in quantity and quality as a vaccine agent against tumors in vitro and in vivo.
Collapse
Affiliation(s)
- Gang Liu
- Department of Surgery, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang 050017, China
- Department of Surgery, Hebei General Hospital, 348 Heping West Road, Shijiazhuang 050051, China
| | - Xiaoyan Fan
- Department of Oncology, Hebei General Hospital, 348 Heping West Road, Shijiazhuang 050051, China
| | - Ying Cai
- Department of Research and Development, Hebei Engineering Technology Research Center for Cell Therapy, Hebei HOFOY Biotech Corporation Ltd, 238 Changjiang Aveneu, Shijiazhuang 500350, China
| | - Zexian Fu
- Department of Surgery, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang 050017, China
| | - Fei Gao
- Department of Surgery, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang 050017, China
| | - Jiantao Dong
- Department of Surgery, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang 050017, China
- Department of Surgery, Hebei General Hospital, 348 Heping West Road, Shijiazhuang 050051, China
| | - Kang Li
- Department of Surgery, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang 050017, China
| | - Jianhui Cai
- Department of Surgery, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang 050017, China
- Department of Surgery, Hebei General Hospital, 348 Heping West Road, Shijiazhuang 050051, China
- Department of Oncology, Hebei General Hospital, 348 Heping West Road, Shijiazhuang 050051, China
| |
Collapse
|
2
|
Taborska P, Bartunkova J, Smrz D. Simultaneous in vitro generation of human CD34 +-derived dendritic cells and mast cells from non-mobilized peripheral blood mononuclear cells. J Immunol Methods 2018; 458:63-73. [PMID: 29684429 DOI: 10.1016/j.jim.2018.04.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/17/2017] [Accepted: 04/18/2018] [Indexed: 10/17/2022]
Abstract
Dendritic cells (DCs) and mast cells (MCs) are key players of the immune system, often coming in close proximity in peripheral tissues. The interplay of these cells is, however, still poorly understood, especially with regards to human cells. The reason for that is the absence of a well established in vitro human cell-based study system that would allow a simultaneous preparation of both cell types. In this study, we show a method for simultaneous generation of DCs and MCs from CD34+ stem cell progenitors that were isolated from the non-adherent fraction of non-mobilized peripheral blood mononuclear cells of healthy donors. We observed that combining stem cells factor (SCF), IL-3 and GM-CSF in serum-free StemPro-34 medium allowed CD34+ cells isolated from an equivalent of 450 ml of peripheral blood to expand to 10-92 × 106 cells after 7 weeks of culturing. These cultures comprised of 6-53% of DCs and 1-21% of MCs as determined by the expression of, respectively, CD11c/HLA-DR or CD117/FcεRI. The DCs were CD1a-CD14-, did not express co-stimulatory molecules CD80 and CD83 and chemokine receptor CCR7. However, the DCs expressed co-stimulatory molecule CD86, and had a capacity to uptake dextran, phagocyte latex particles and induce alloreactivity. MCs, on the other hand, degranulated after crosslinking of FcεRI-bound IgE as determined by the externalization of CD107b. Collectively, our data show that CD34+-derived human DCs and MCs can be generated in a single culture using CD34+ cells isolated from non-mobilized human peripheral blood and that this method may allow ex vivo studies on DC-MC interplay in human system.
Collapse
Affiliation(s)
- Pavla Taborska
- Institute of Immunology, Charles University, 2nd Faculty of Medicine, University Hospital Motol, Czech Republic
| | - Jirina Bartunkova
- Institute of Immunology, Charles University, 2nd Faculty of Medicine, University Hospital Motol, Czech Republic
| | - Daniel Smrz
- Institute of Immunology, Charles University, 2nd Faculty of Medicine, University Hospital Motol, Czech Republic.
| |
Collapse
|
3
|
MENG FANDONG, WANG SHUAI, JIANG YOUHONG, SUI CHENGGUANG. Antitumor effect of dendritic cells transfected with prostate-specific membrane antigen recombinant adenovirus on prostate cancer: An in vitro study. Mol Med Rep 2016; 13:2124-34. [DOI: 10.3892/mmr.2016.4754] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 09/25/2015] [Indexed: 11/06/2022] Open
|
4
|
Kumar J, Kale V, Limaye L. Umbilical cord blood-derived CD11c(+) dendritic cells could serve as an alternative allogeneic source of dendritic cells for cancer immunotherapy. Stem Cell Res Ther 2015; 6:184. [PMID: 26407613 PMCID: PMC4583174 DOI: 10.1186/s13287-015-0160-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 08/17/2015] [Accepted: 08/17/2015] [Indexed: 11/10/2022] Open
Abstract
Introduction Allogenic dendritic cells (DCs) generated from healthy donors, who are complete or partially HLA-matched, have been used for clinical trials. One of the sources for allogenic DCs is umbilical cord blood (UCB) cells. However, as far as cord blood cells are concerned, looking at their naïve nature, there is a concern as to whether the DCs generated from them will have enough potential to elicit a proper T cell response. For this, we compared CD11c+ UCB-DCs/ Cytotoxic T lymphocytes (CTLs) with the conventional source, i.e. peripheral blood (PBL) monocyte DCs/CTLs, using various parameters. Methods CD11c+ DCs generated from the two sources were compared morphologically, phenotypically and functionally. Functional assays included antigen uptake, chemotactic migration and MLR (mixed lymphocyte reaction). The CTLs generated were examined for the activation markers, granzyme A & granzyme B, and IFN-γ secretion. MUC1 (STAPPVHNV) peptide-specific CTLs were quantified by Streptamer staining. In vitro CTL activity was assessed by their efficiency in killing MCF-7 cells. For in vivo CTL assay, a xenograft of MCF-7-luc-F5 cells in female NOD/SCID mice was employed. Regression of tumors in mice was monitored using an in vivo imaging system before and after ten days of CTL infusion. Statistical analysis of all the experiments between the two groups was evaluated by one-way ANOVA. Results The CD11c+ DCs from the two sources were morphologically and phenotypically similar. Their capacity to uptake antigen, migration towards CCL-19 and MLR activity were equivalent. UCB-CTLs had significantly higher levels of activation markers, number of MUC1 specific CTLs, IFN-γ secretion and IL-12p70/IL-10 ratio than that of PBL-CTLs. Hematoxylin and Eosin-stained tumor sections showed T cell infiltration, which was further confirmed by immunofluorescence staining. In vivo CTL activity was found to be similar with the two sources. Conclusions Our data demonstrate that CD11c+ UCB-DCs/CTLs are as potent as standard CD11c+ PBL-DC/CTLs and could therefore be used as an allogenic source for therapeutic purposes. The findings of this study could help in taking us one step closer towards the personalized therapy using DC based cancer vaccines. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0160-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jeetendra Kumar
- Stem Cell Laboratory, National Centre for Cell Science, Ganeshkhind, Pune, 411007, India.
| | - Vaijayanti Kale
- Stem Cell Laboratory, National Centre for Cell Science, Ganeshkhind, Pune, 411007, India.
| | - Lalita Limaye
- Stem Cell Laboratory, National Centre for Cell Science, Ganeshkhind, Pune, 411007, India.
| |
Collapse
|
5
|
Pham VQ, Nguyen ST, Van Pham P. Production of functional dendritic cells from mouse bone marrow. BIOMEDICAL RESEARCH AND THERAPY 2014. [DOI: 10.7603/s40730-014-0020-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
6
|
Fan J, Cai H, Li Q, Du Z, Tan W. The effects of ROS-mediating oxygen tension on human CD34+CD38− cells induced into mature dendritic cells. J Biotechnol 2012; 158:104-11. [DOI: 10.1016/j.jbiotec.2012.01.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 01/04/2012] [Accepted: 01/14/2012] [Indexed: 11/29/2022]
|
7
|
Chiang CLL, Hagemann AR, Leskowitz R, Mick R, Garrabrant T, Czerniecki BJ, Kandalaft LE, Powell DJ, Coukos G. Day-4 myeloid dendritic cells pulsed with whole tumor lysate are highly immunogenic and elicit potent anti-tumor responses. PLoS One 2011; 6:e28732. [PMID: 22194898 PMCID: PMC3237492 DOI: 10.1371/journal.pone.0028732] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 11/14/2011] [Indexed: 11/25/2022] Open
Abstract
“Day-7” myeloid DCs are commonly used in the clinic. However, there is a strong need to develop DCs faster that have the same potent immunostimulatory capacity as “Day-7” myeloid DCs and at the same time minimizing time, labor and cost of DC preparations. Although “2 days” DCs can elicit peptide-specific responses, they have not been demonstrated to engulf, process and present complex whole tumor lysates, which could be more convenient and personalized source of tumor antigens than defined peptides. In this preclinical study, we evaluated the T-cell stimulatory capacity of Day-2, Day-4, and Day-7 cultured monocyte-derived DCs loaded with SKOV3 cell whole lysate prepared by freeze-thaw or by UVB-irradiation followed by freeze-thaw, and matured with lipopolysaccharide (LPS) and interferon (IFN)-gamma. DCs were evaluated for antigen uptake, and following maturation with LPS and IFN-gamma, DCs were assessed for expression of CD80, CD40, CD86, ICAM-1 and CCR7, production of IL-12p70 and IP-10, and induction of tumor-specific T-cell responses. Day-4 and Day-7 DCs exhibited similar phagocytic abilities, which were superior to Day-2 DCs. Mature Day-7 DCs expressed the highest CD40 and ICAM-1, but mature Day-4 DCs produced the most IL-12p70 and IP-10. Importantly, Day-4 and Day-7 DCs derived from ovarian cancer patients stimulated equally strongly tumor-specific T-cell responses. This is the first study demonstrating the highly immunogenic and strong T-cell stimulatory properties of Day-4 myeloid DCs, and provided important preclinical data for rapid development of potent whole tumor lysate-loaded DC vaccines that are applicable to many tumor types.
Collapse
Affiliation(s)
- Cheryl Lai-Lai Chiang
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Andrea R. Hagemann
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Rachel Leskowitz
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Rosemarie Mick
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Thomas Garrabrant
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Brian J. Czerniecki
- Rena Rowan Breast Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Lana E. Kandalaft
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Daniel J. Powell
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - George Coukos
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
8
|
Abstract
Stem and progenitor cells are present in cord blood at a high frequency making these cells a major target population for experimental and clinical studies. Over the past decade there has been considerable developments in cord blood research and transplantation but despite the rapid progress many problems remain. The initial hope that cord blood would be an alternative source of haemopoietic cells for transplantation has been tempered by the fact that there are insufficient cells in most cord blood collections to engraft an adult of average weight. In attempts to increase the cell number, a plethora of techniques for ex-vivo expansion have been developed.These techniques have also proved useful for gene therapy. As cord blood cells possess unique properties this allows them to be utilised as suitable vehicles for gene therapy and long-term engraftment of transduced cells has been achieved. Current work examining the nature of the stem cells present in this haematological source indicates that cord blood contains not only haemopoietic stem cells but also primitive non-haemopoietic cells with high proliferative and developmental potential. As attention focuses on stem cell biology and the controversies surrounding the potential use of embryonic stem cells in treatment of disease, the properties of stem cells from other sources including cord blood are being re-appraised. The purpose of this article is to review some of the current areas of work and highlight biological problems associated with the use of cord blood cells.
Collapse
Affiliation(s)
- E A de Wynter
- Molecular Medicine Unit, University of Leeds, St. James's University Hospital, Leeds, U.K. (E-mail,
| |
Collapse
|
9
|
Phuc PV, Lam DH, Ngoc VB, Thu DT, Nguyet NTM, Ngoc PK. Production of functional dendritic cells from menstrual blood--a new dendritic cell source for immune therapy. In Vitro Cell Dev Biol Anim 2011; 47:368-75. [PMID: 21424240 DOI: 10.1007/s11626-011-9399-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 02/22/2011] [Indexed: 12/23/2022]
Abstract
Dendritic cells (DCs) are the most professional antigen-presenting cells of the mammalian immune system. They are able to phagocytize, process antigen materials, and then present them to the surface of other cells including T lymphocytes in the immune system. These capabilities make DC therapy become a novel and promising immune-therapeutic approach for cancer treatment as well as for cancer vaccination. Many trials of DC therapy to treat cancers have been performed and have shown their application value. They involve harvesting monocytes or hematopoietic stem cells from a patient and processing them in the laboratory to produce DCs and then reintroduced into a patient in order to activate the immune system. DCs were successfully produced from peripheral, umbilical cord blood-derived monocytes or hematopoietic stem cells. In this research, we produced DCs from human menstrual blood-derived monocytes. Briefly, monocytes were isolated by FACS based on FSC vs. SSC plot from lysed menstrual blood. Obtained monocytes were induced into DCs by a two-step protocol. In the first step, monocytes were incubated in RPMI medium supplemented with 2% FBS, GM-CSF, and IL-4, followed by incubation in RPMI medium supplemented with α-TNF in the second step. Our data showed that induced monocytes had typical morphology of DCs, expressed HLA-DR, HLA-ABC, CD80 and CD86 markers, exhibited uptake of dextran-FITC, stimulated allogenic T cell proliferation, and released IL-12. These results demonstrated that menstrual blood can not only be a source of stromal stem cell but also DCs, which are a potential candidate for immune therapy.
Collapse
Affiliation(s)
- Pham Van Phuc
- Laboratory of Stem cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh, Vietnam.
| | | | | | | | | | | |
Collapse
|
10
|
Balan S, Kale VP, Limaye LS. A large number of mature and functional dendritic cells can be efficiently generated from umbilical cord blood-derived mononuclear cells by a simple two-step culture method. Transfusion 2011; 50:2413-23. [PMID: 20497510 DOI: 10.1111/j.1537-2995.2010.02706.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Advances in the past two decades in dendritic cell (DC) biology paved the way to exploit them as a promising tool in cancer immunotherapy. The prerequisite for DC vaccine preparations is large-scale in vitro generations of homogeneous, mature, and functional DCs. Frequent improvements are being made in the existing in vitro DC production protocols to achieve this goal. In our previous study we reported a large-scale generation of mature, functional DCs from umbilical cord blood (UCB) CD34+ cells. Here we report that this method can be used for the efficient generation of DCs from UCB mononuclear cells (MNCs) and thus the hematopoietic stem cell isolation step is not essential. STUDY DESIGN AND METHODS MNCs or CD34+ cells isolated from the same cord blood (CB) samples were used for the generation of DCs. DCs were characterized for morphology, phenotype, and functional assays including antigen uptake, chemotaxis, and mixed leukocyte reaction. Similarly DCs generated from the MNCs of same fresh and frozen CB units were compared. RESULTS The morphologic, phenotypic, and functional characterization of the DCs generated from various sets show that they were comparable in nature irrespective of the starting population used. CONCLUSION We conclude that the CD34+ isolation step is not essential for the generation of mature, functional DCs and thus can be eliminated. More importantly, we show that DCs can be generated with equal efficiency from the MNCs of frozen CB units. Our culture method will be useful for exploiting the potential of UCB as an additional source for allogeneic DCs in the clinical settings.
Collapse
Affiliation(s)
- Sreekumar Balan
- National Centre for Cell Science, Pune University Campus, Pune, India
| | | | | |
Collapse
|
11
|
Ex vivo development, expansion and in vivo analysis of a novel lineage of dendritic cells from hematopoietic stem cells. JOURNAL OF IMMUNE BASED THERAPIES AND VACCINES 2010; 8:8. [PMID: 21106069 PMCID: PMC3004889 DOI: 10.1186/1476-8518-8-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Accepted: 11/24/2010] [Indexed: 12/11/2022]
Abstract
Dendritic cells (DCs) play a key role in innate and adaptive immunity but the access to sufficient amount of DCs for basic and translational research has been limited. We established a novel ex vivo system to develop and expand DCs from hematopoietic stem/progenitor cells (HPCs). Both human and mouse HPCs were expanded first in feeder culture supplemented with c-Kit ligand (KL, stem cell factor, steel factor or CD117 ligand), Flt3 ligand (fms-like tyrosine kinase 3, Flt3L, FL), thrombopoietin (TPO), IL-3, IL-6, and basic fibroblast growth factor (bFGF), and then in a second feeder culture ectopically expressing all above growth factors plus GM-CSF and IL-15. In the dual culture system, CD34+ HPCs differentiated toward DC progenitors (DCPs), which expanded more than five orders of magnitude. The DCPs showed myeloid DC surface phenotype with up-regulation of transcription factors PU.1 and Id2, and DC-related factors homeostatic chemokine ligand 17 (CCL17) and beta-chemokine receptor 6 (CCR6). Multiplex ELISA array and cDNA microarray analyses revealed that the DCPs shared some features of IL-4 and IL-15 DCs but displayed a pronounced proinflammatory phenotype. DCP-derived DCs showed antigen-uptake and immune activation functions analogous to that of the peripheral blood-derived DCs. Furthermore, bone marrow HPC-derived DCP vaccines of tumor-bearing mice suppressed tumor growth in vivo. This novel approach of generating DCP-DCs, which are different from known IL-4 and IL-15 DCs, overcomes both quantitative and qualitative limitations in obtaining functional autologous DCs from a small number of HPCs with great translational potential.
Collapse
|
12
|
Balan S, Kale VP, Limaye LS. A simple two-step culture system for the large-scale generation of mature and functional dendritic cells from umbilical cord blood CD34+ cells. Transfusion 2009; 49:2109-21. [DOI: 10.1111/j.1537-2995.2009.02231.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
13
|
Abe T, Fuse I, Narita M, Takahashi M, Aizawa Y. Combination use of immune complexes and a Ca2(+) channel blocker azelnidipine enhances interleukin-12 p40 secretion without T helper type 17 cytokine secretion in human monocyte-derived dendritic cells. Clin Exp Immunol 2009; 156:405-12. [PMID: 19438591 DOI: 10.1111/j.1365-2249.2009.03911.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Immune complexes (ICs) improve the capacity of priming specific CD8(+) cytotoxic T cell responses of dendritic cells (DCs). ICs induce phosphorylation of mitogen-activated protein kinases (MAPK) and calcium influx, although the precise regulating mechanism still remains unclear. In the present study, we investigated the effect of a Ca2(+) channel blocker on the phosphorylation of p38 MAPK and extracellular signal-regulated kinase (ERK) in immature monocyte-derived DCs stimulated with lipopolysaccharide (LPS) or LPS-ICs, and the production of interleukin (IL)-12 family members (p40, p70, IL-23), T helper type 17 (Th17) cytokines (IL-6 and IL-23), tumour necrosis factor (TNF)-alpha and IL-10 were also investigated. In comparison with LPS stimulation, LPS-ICs stimulation enhanced p38 MAPK phosphorylation significantly, which was associated with an increase in IL-12 p40 monomer/homodimer secretion. LPS-ICs also enhanced TNF-alpha and IL-6 secretion, but suppressed IL-23 secretion. The use of azelnidipine (Aze), a long-acting L-type Ca2(+) channel blocker with a high lipid solubility, suppressed p38 MAPK phosphorylation stimulated with LPS or LPS-ICs, but surprisingly enhanced IL-12 p40 monomer/homodimer secretion stimulated with LPS-ICs. This IL-12 p40 secretion-enhancing effect was not accompanied by IL-10 or IL-23 production, but was associated with ERK phosphorylation. The use of Aze did not affect IL-12 p70 production. These results suggest that the use of Aze enhances ICs-mediated IL-12 p40 secretion without additional IL-23 secretion. Therefore, the use of Aze and ICs could be a new therapeutic approach to immunomolecular therapy, as it does not cause Th17 differentiation which induces autoimmunity or reduces anti-tumour immunity.
Collapse
Affiliation(s)
- T Abe
- Niigata University Graduate School of Medicine and Dental Science, Division of Hematology, Department of Regenerative and Transplant Medicine, Niigata, Japan.
| | | | | | | | | |
Collapse
|
14
|
Abstract
Dendritic cells (DCs) ‘pulsed’ with an appropriate antigen may elicit an antitumour immune response in mouse models. However, while attempting to develop a DC immunotherapy protocol for the treatment of breast cancer based on the tumour-associated MUC1 glycoforms, we found that unpulsed DCs can affect tumour growth. Protection from RMA-MUC1 tumour challenge was achieved in C57Bl/6 MUC1 transgenic mice by immunising with syngeneic DCs pulsed with a MUC1 peptide. However, unpulsed DCs gave a similar level of protection, making it impossible to evaluate the effect of immunisation of mice with DCs pulsed with the specific peptide. Balb/C mice could also be protected from tumour challenge by immunisation with unpulsed DCs prior to challenge with murine mammary tumour cells (410.4) or these cells transfected with MUC1 (E3). Protection was achieved with as few as three injections of 50 000 naïve DCs per mouse per week, was not dependent on injection route, and was not specific to cell lines expressing human MUC1. However, the use of Rag2-knockout mice demonstrated that the adaptive immune response was required for tumour rejection. Injection of unpulsed DCs into mice bearing the E3 tumour slowed tumour growth. In vitro, production of IFN-γ and IL-4 was increased in splenic cells isolated from mice immunised with DCs. Depleting CD4 T cells in vitro partially decreased cytokine production by splenocytes, but CD8 depletion had no effect. This paper shows that naïve syngeneic DCs may induce an antitumour immune response and has implications for DC immunotherapy preclinical and clinical trials.
Collapse
|
15
|
Lin VK, Wang SY, Vazquez DV, C Xu C, Zhang S, Tang L. Prostatic stromal cells derived from benign prostatic hyperplasia specimens possess stem cell like property. Prostate 2007; 67:1265-76. [PMID: 17597114 DOI: 10.1002/pros.20599] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION The hyper-proliferative activity of stromal smooth muscle (SM) cells is believed to be responsible for the pathogenesis of benign prostatic hyperplasia (BPH). We have observed that those stromal cells can differentiate into unrelated specialized cells. We thus hypothesize that stromal cells derived from adults prostate specimens may contain adult stem cells. To test this hypothesis, human prostate stromal primary cultures were established and used for characterization of their stem cell properties. METHODS Immunoblotting, immunohistochemistry, RT-PCR, and tissue culture techniques were used to characterize the primary cultured human prostate-derived stromal cells for their stem cell and differentiation properties. The plasticity of these stromal cells was analyzed using cell culture and histology techniques. RESULTS Primary cultured prostate stromal cells from BPH patient possess polygonal and elongated fibroblast/myofibroblast cellular morphology. They are positive in CD30, CD34, CD44, NSE, CD133, Flt-1, stem cell factor (SCF), and neuron-specific enolase (NSE), but negative in C-Kit, stem cell antigen (SCA), SH2, CD11b. Expression of SM myogenic markers in these cells may be induced by sodium butyrate (NaBu) treatment. Induction to osteogenic and adipogenic differentiation in these cells is also evident. CONCLUSIONS Our study on primary stromal cells from BPH patients have yielded many interesting findings that these prostate stroma cells possess: (1) mesenchymal stem cell (MSC) markers; (2) strong proliferative potential; and (3) ability to differentiate or transdifferentiate to myogenic, adipogenic, and osteogenic lineages. These cell preparations may serve as a potential tool for studies in prostate adult stem cell research and the regulation of benign prostatic hyperplasia.
Collapse
Affiliation(s)
- Victor K Lin
- Department of Urology, The University of Texas, Southwestern Medical Center, Dallas, Texas 75390-9110, USA.
| | | | | | | | | | | |
Collapse
|
16
|
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
|
17
|
Slukvin II, Vodyanik MA, Thomson JA, Gumenyuk ME, Choi KD. Directed differentiation of human embryonic stem cells into functional dendritic cells through the myeloid pathway. THE JOURNAL OF IMMUNOLOGY 2006; 176:2924-32. [PMID: 16493050 DOI: 10.4049/jimmunol.176.5.2924] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We have established a system for directed differentiation of human embryonic stem (hES) cells into myeloid dendritic cells (DCs). As a first step, we induced hemopoietic differentiation by coculture of hES cells with OP9 stromal cells, and then, expanded myeloid cells with GM-CSF using a feeder-free culture system. Myeloid cells had a CD4+CD11b+CD11c+CD16+CD123(low)HLA-DR- phenotype, expressed myeloperoxidase, and included a population of M-CSFR+ monocyte-lineage committed cells. Further culture of myeloid cells in serum-free medium with GM-CSF and IL-4 generated cells that had typical dendritic morphology; expressed high levels of MHC class I and II molecules, CD1a, CD11c, CD80, CD86, DC-SIGN, and CD40; and were capable of Ag processing, triggering naive T cells in MLR, and presenting Ags to specific T cell clones through the MHC class I pathway. Incubation of DCs with A23187 calcium ionophore for 48 h induced an expression of mature DC markers CD83 and fascin. The combination of GM-CSF with IL-4 provided the best conditions for DC differentiation. DCs obtained with GM-CSF and TNF-alpha coexpressed a high level of CD14, and had low stimulatory capacity in MLR. These data clearly demonstrate that hES cells can be used as a novel and unique source of hemopoietic and DC precursors as well as DCs at different stages of maturation to address essential questions of DC development and biology. In addition, because ES cells can be expanded without limit, they can be seen as a potential scalable source of cells for DC vaccines or DC-mediated induction of immune tolerance.
Collapse
Affiliation(s)
- Igor I Slukvin
- Department of Pathology and Laboratory Medicine, Wisconsin National Primate research Center, University of Wisconsin, Madison, WI 53706, USA.
| | | | | | | | | |
Collapse
|
18
|
Hieronymus T, Gust TC, Kirsch RD, Jorgas T, Blendinger G, Goncharenko M, Supplitt K, Rose-John S, Müller AM, Zenke M. Progressive and Controlled Development of Mouse Dendritic Cells from Flt3+CD11b+Progenitors In Vitro. THE JOURNAL OF IMMUNOLOGY 2005; 174:2552-62. [PMID: 15728461 DOI: 10.4049/jimmunol.174.5.2552] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Dendritic cells (DC) represent key regulators of the immune system, yet their development from hemopoietic precursors is poorly defined. In this study, we describe an in vitro system for amplification of a Flt3(+)CD11b(+) progenitor from mouse bone marrow with specific cytokines. Such progenitor cells develop into both CD11b(+) and CD11b(-) DC, and CD8alpha(+) and CD8alpha(-) DC in vivo. Furthermore, with GM-CSF, these progenitors synchronously differentiated into fully functional DC in vitro. This two-step culture system yields homogeneous populations of Flt3(+)CD11b(+) progenitor cells in high numbers and allows monitoring the consecutive steps of DC development in vitro under well-defined conditions. We used phenotypic and functional markers and transcriptional profiling by DNA microarrays to study the Flt3(+)CD11b(+) progenitor and differentiated DC. We report here on an extensive analysis of the surface Ag expression of Flt3(+)CD11b(+) progenitor cells and relate that to surface Ag expression of hemopoietic stem cells. Flt3(+)CD11b(+) progenitors studied exhibit a broad overlap of surface Ags with stem cells and express several stem cell Ags such as Flt3, IL-6R, c-kit/SCF receptor, and CD93/AA4.1, CD133/AC133, and CD49f/integrin alpha(6). Thus, Flt3(+)CD11b(+) progenitors express several stem cell surface Ags and develop into both CD11b(+) and CD11b(-) DC, and CD8alpha(+) and CD8alpha(-) DC in vivo, and thus into both of the main conventional DC subtypes.
Collapse
Affiliation(s)
- Thomas Hieronymus
- Institute for Biomedical Engineering-Cell Biology, University Medical School Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Xu RLC, Tang Y, Ogburn PL, Malinowski K, Madajewicz S, Santiago-Schwarz F, Fan Q. Implication of delayed TNF-α exposure on dendritic cell maturation and expansion from cryopreserved cord blood CD34+ hematopoietic progenitors. J Immunol Methods 2004; 293:169-82. [PMID: 15541286 DOI: 10.1016/j.jim.2004.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2004] [Revised: 07/30/2004] [Accepted: 08/03/2004] [Indexed: 10/26/2022]
Abstract
Most currently used systems for dendritic cell (DC) production from progenitors entail tumor necrosis factor alpha (TNF-alpha) at the onset of cell culture, based on the notion that TNF-alpha might be required in the early stages of DC development. To optimize conditions for DC expansion from cryopreserved cord blood (CB) CD34+ hematopoietic progenitors, we took a dynamic approach to define the timing of TNF-alpha exposure to the culture. We cultured cord blood CD34+ cells in RPMI-1640 with 10% human AB plasma, stem cell factor (days 1-6), granulocyte-macrophage colony-stimulating factor (days 1-18), interleukin-4 (days 6-18) and varying schedules of TNF-alpha (0-144 h after thawing). Expression of the DC-associated markers, including CD83/CD1a, HLA DR/CD86/CD80, CD14/CD40, was monitored every 3 days. Our data demonstrate that delayed TNF-alpha exposure by 48-72 h after thawing gave rise to two- to three-fold increase in the yield of CD83+ DCs that were highly active in stimulating allogeneic T-cell proliferation compared to immediate TNF-alpha exposure. Thus, the immediate exposure of cryopreserved cord blood CD34+ cells to TNF-alpha, potentially compromising DC expansion, should be avoided. This finding should be of significant consideration when using cryopreserved CD34+ progenitor cells as a source of immunologically competent DCs in a clinical setting.
Collapse
Affiliation(s)
- Richard Li-Cheng Xu
- Department of Medicine and Long Island Cancer Institute, State University of New York at Stony Brook, NY, USA.
| | | | | | | | | | | | | |
Collapse
|
20
|
Diwan M, Elamanchili P, Lane H, Gainer A, Samuel J. Biodegradable nanoparticle mediated antigen delivery to human cord blood derived dendritic cells for induction of primary T cell responses. J Drug Target 2004; 11:495-507. [PMID: 15203918 DOI: 10.1080/10611860410001670026] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Dendritic cells (DCs) in the peripheral tissues act as sentinels of the immune system. They detect and capture pathogens entering the body and present their antigens to T cells to trigger responses directed towards elimination of the pathogen. The induction of peripheral tolerance against self and certain foreign antigens is also believed to be mediated through DCs. The outcome of any immune response is largely controlled by the microenvironment of antigen capture, processing and presentation by DCs. The "context" of antigen delivery to DCs will directly influence the microenvironment of antigen presentation and hence the regulation of immune responses. We report here preliminary investigations describing the formulation of a pharmaceutically acceptable, biodegradable, and strategic nanoparticulate delivery system, and its application for efficient antigen loading of DCs to achieve antigen specific T cell activation. "Pathogen-mimicking" nanoparticles capable of interacting with DCs were fabricated by incorporating monophosphoryl lipid A (MPLA; toll-like receptor (TLR) 4 ligand) or CpG ODN (seq #2006; TLR9 ligand) in biodegradable copolymer, poly(D,L,-lactic-co-glycolic acid) (PLGA). The uptake of PLGA nanoparticles by human umbilical cord blood derived DCs (DCs propagated from CD34 progenitors) was conclusively demonstrated by scanning electron microscopy (SEM), fluorescence activated cell sorting (FACS) and confocal laser scanning microscopy (CLSM). Cell phenotype at day 12 of cultures was determined as immature DC using specific cell surface markers, i.e. CD11c (approximately 90%), MHC-II (approximately 70%), CD86 (approximately 20%), CD83 (approximately 5%), CD80 (approximately 40%), CD40 (approximately 40%), and CCR7 (approximately 5%). Tetanus toxoid (TT), a model antigen, was encapsulated in nanoparticles along with an immunomodulator, i.e. either MPLA or CpG ODN. DCs pulsed with various antigen formulations were co-cultured with autologous naïve T cells at various cell ratios (DC: T cells were 1:5-20). The DCs pulsed with TT and MPLA together in nanoparticles induced significantly higher T cell proliferation (P<0.05) as compared to when DCs pulsed with TT and MPLA in solution were employed. A similar trend was observed when CpG ODN was used instead of MPLA in the TT nanoparticles. This strategy of antigen delivery to DCs was then tested with a cancer vaccine candidate, a MUC1 lipopeptide. The T cell proliferation observed in the presence of nanoparticulate MUC1 and MPLA pulsed-DCs was much higher than DCs pulsed with soluble antigen (P<0.0005). These results indicate that PLGA nanoparticles mimicking certain features of pathogens are efficient delivery systems for targeting vaccine antigens to DCs and activation of potent T cell responses.
Collapse
Affiliation(s)
- Manish Diwan
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | | | | | | | | |
Collapse
|
21
|
Morse MA, Chui S, Clay TM, Lyerly HK. Recent areas of development for dendritic cell vaccines. ACTA ACUST UNITED AC 2004; 21:339-50. [PMID: 15338754 DOI: 10.1016/s0921-4410(03)21017-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
|
22
|
Navarrete CV, Gómez J, Borràs FE. Cord blood dendritic cells: subsets, functional characteristics and in vitro generation. Leuk Lymphoma 2003; 44:923-8. [PMID: 12854889 DOI: 10.1080/1042819031000068070] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dendritic cells (DCs) form a heterogeneous population of cells capable of stimulating naive T cells and initiating primary immune responses. This well-known function of DCs has offered the possibility of developing clinical protocols for their use in immunotherapy to tumours. DCs may also play a critical role in the induction of peripheral immunological tolerance, which could have important implications in the treatment of autoimmunity or in the outcome of clinical transplantation. Recent reports have indicated that cord blood transplantation is associated with a reduced incidence of graft versus host disease. Thus studies on the identification and characterisation of DCs present in, or derived from cord blood will help to understand their role, not only in neonatal immunity, but also in the outcome of cord blood transplantation.
Collapse
Affiliation(s)
- Cristina V Navarrete
- Department of Histocompatibility and Immunogenetics, National Blood Service, Colindale Avenue, London NW9 5BG, UK.
| | | | | |
Collapse
|