Báo cáo hóa học: " Molecular signatures of maturing dendritic cells: implications for testing the quality of dendritic cell therapies" pptx

15 499 0
Báo cáo hóa học: " Molecular signatures of maturing dendritic cells: implications for testing the quality of dendritic cell therapies" pptx

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

Jin et al Journal of Translational Medicine 2010, 8:4 http://www.translational-medicine.com/content/8/1/4 RESEARCH Open Access Molecular signatures of maturing dendritic cells: implications for testing the quality of dendritic cell therapies Ping Jin1*†, Tae Hee Han1,2†, Jiaqiang Ren1, Stefanie Saunders1, Ena Wang1, Francesco M Marincola1, David F Stroncek1 Abstract Background: Dendritic cells (DCs) are often produced by granulocyte-macrophage colony-stimulating factor (GMCSF) and interleukin-4 (IL-4) stimulation of monocytes To improve the effectiveness of DC adoptive immune cancer therapy, many different agents have been used to mature DCs We analyzed the kinetics of DC maturation by lipopolysaccharide (LPS) and interferon-g (IFN-g) induction in order to characterize the usefulness of mature DCs (mDCs) for immune therapy and to identify biomarkers for assessing the quality of mDCs Methods: Peripheral blood mononuclear cells were collected from healthy subjects by apheresis, monocytes were isolated by elutriation, and immature DCs (iDCs) were produced by days of culture with GM-CSF and IL-4 The iDCs were sampled after 4, and 24 hours in culture with LPS and IFN-g and were then assessed by flow cytometry, ELISA, and global gene and microRNA (miRNA) expression analysis Results: After 24 hours of LPS and IFN-g stimulation, DC surface expression of CD80, CD83, CD86, and HLA Class II antigens were up-regulated Th1 attractant genes such as CXCL9, CXCL10, CXCL11 and CCL5 were up-regulated during maturation but not Treg attractants such as CCL22 and CXCL12 The expression of classical mDC biomarker genes CD83, CCR7, CCL5, CCL8, SOD2, MT2A, OASL, GBP1 and HES4 were up-regulated throughout maturation while MTIB, MTIE, MTIG, MTIH, GADD45A and LAMP3 were only up-regulated late in maturation The expression of miR-155 was up-regulated 8-fold in mDCs Conclusion: DCs, matured with LPS and IFN-g, were characterized by increased levels of Th1 attractants as opposed to Treg attractants and may be particularly effective for adoptive immune cancer therapy Introduction Dendritic cells (DC) are key players in both innate and adaptive immune responses They are potent antigen presenting cells that recognize, process, and present antigens to T-cells in vivo [1-3] Consequently, DC-based immunotherapy has become one of the most promising approaches for the treatment of cancer [4,5] The frequency of DCs in the peripheral blood is naturally low and they are difficult to separate from other peripheral blood leukocytes [6], therefore, to enhance DC function, hematopoietic progenitor cells or peripheral blood * Correspondence: pjin@cc.nih.gov † Contributed equally Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA monocytes are usually used to produce mDC in vitro by culture with growth factors and cytokines [6,7] Large quantities of mononuclear cells can easily be collected from the peripheral blood by leukapheresis Monocytes can be isolated from other leukocytes collected by apheresis with high purity by adherence, elutriation, or using immunomagnetic beads [8-10] To produce immature DCs (iDCs), monocytes are usually incubated with granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-4 (IL-4) Because mature DCs (mDCs) are superior to iDCs for the stimulation of cytotoxic T-cells, iDCs derived from monocytes are often treated with various exogenous stimuli known to induce DCs maturation including lipopolysaccharide (LPS) and interferon-g (IFN-g) [5,11] One of the goals © 2010 Jin et al; licensee BioMed Central Ltd This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited Jin et al Journal of Translational Medicine 2010, 8:4 http://www.translational-medicine.com/content/8/1/4 of this study was to characterize the molecular profile of changes associated with LPS and IFN-g induced DC maturation to estimate the effectiveness of these mDCs in adoptive immune cancer therapy When developing cellular therapies such as mDCs it is often necessary to compare products manufactured with a standard method and an alternative method It is also necessary to determine if products manufactured from the starting material of different people are consistent or similar Once the manufacturing process has been established and clinical products are being manufactured, clinical cellular therapies must also be assessed for potency Another goal of this study was to identify molecular biomarkers that were associated with DC maturation and in order to characterize mDCs and that could be used for consistency, comparibility, and potency testing DCs are often assessed by flow cytometry for the expression of the costimulatory molecules CD80 and CD86, the maturation marker CD83, the chemokine receptor CCR7, and antigen presentation molecules, HLA class II antigens, to document the transition of iDCs to mDCs Some cellular therapy laboratories also test the function of DCs by measuring their ability to produce IL-12, IL-10, IL-23 or IFN-g following stimulation However, the diverse functions of DC therapies indicate that additional biomarkers are necessory to characterize mDCs Based on the multiple functions of DCs and their broad spectrum of effector molecules, it is highly improbable that a limited number of biomarkers can adequately measure DC potency But whole transcriptome expression analysis and microRNA (miR) profiling analysis of the DC maturation process could provide better insight into DC biology and identify biomarkers that are indicators of DC potency Although monocytes, iDCs, and mDCs have been characterized at a molecular level, few studies have comprehensively studied the molecular events associated with DC maturation In this study we compared the kinetics of global changes of both gene and miR expression associated with LPS and IFN-g induced DC maturation Gene and miR changes in DCs were assessed after 4, and 24 hours of LPS and IFN-g stimulation To validate the functional activity of DCs, we also tested soluble protein production in culture supernatant after 24 hours of maturation and after incubation with CD40 ligand transfected mouse fibroblasts Materials and methods Study design Peripheral blood mononuclear cell (PBMC) concentrates were collected using a CS3000 Plus blood cell separator (Baxter Healthcare Corp., Fenwal Division, Page of 15 Deerfield, IL) from healthy donors in the Department of Transfusion Medicine (DTM), Clinical Center, National Institutes of Health (NIH) All donors signed an informed consent approved by a NIH Institutional Review Board Monocytes were isolated from the PBMC concentrates on the day of PBMC collection by elutriation (Elutra®, Gambro BCT, Lakewood, CO) using the instrument’s automatic mode according to the manufacturer’s recommendations The monocytes were treated with GM-CSF (2000 IU/mL, R&D Systems, Minneapolis, MN) and IL-4 (2000 IU/mL, R&D Systems) for days to produce iDCs The iDCs were then treated for 24 hours with LPS and IFN-g to produce mDCs The results of analysis of iDCs and mDCs by flow cytometry and gene expression profiling have been previously published [12] DC preparation, maturation, and harvest The elutriated monocytes from each donor were suspended at 6.7 × 106/mL with RPMI 1640 (Invitrogen, Carlsbad, CA) supplemented with 10% fetal calf serum (FSC) (Invitrogen), mM L-glutamine (Invitrogen), 1% nonessential amino acids (Invitrogen), 1% pyruvate (Invitrogen), 100 units/mL penicillin/streptomycin (Invitrogen), and 50 μM 2-mercaptoethanol (Sigma, St Louis, MO) A total of 10 mL of monocyte suspension was cultured in T25 culture flasks (Nalge Nunc International, Rochester, NY) overnight in a humidified incubator with 5% CO at 37°C On Day 1, 2000 IU/mL human IL-4 (R&D Systems) and 2000 IU/mL GM-CSF (R&D Systems) were added to the culture On Day 3, an additional 2000 IU/mL IL-4 and GM-CSF were added To induce DC maturation, on day 4, 100 ng/mL LPS (Sigma) and 1000 IU/mL IFN-g (R&D Systems) were added The DCs were harvested at 0, 4, and 24 hours (h) after the addition of LPS and IFN-g To remove the adherent DCs, mM EDTA-PBS was added to each flask on ice The harvested cells were pelleted, washed twice with HBSS, and resuspended in RPMI 1640 The total number of cells harvested and their viability was measured microscopically after adding Trypan Blue Flow cytometeric analysis The purity of the elutriated monocytes was evaluated by flow cytometry using CD14-PE, CD19-FITC, CD3-PECy5, and CD56-APC (Becton Dickinson, Mountain View, CA) and isotype controls (Becton Dickinson) To confirm the maturation of the DCs, the harvested DCs were tested with CD80-FITC, CD83-PE, CD86-FITC, HLA-DR-PE-Cy5, and CD14-APC (Becton Dickinson) and isotype controls (Becton Dickson) Flow cytometry acquisition and analysis were performed with a FACScan using CellQuest software (Becton Dickinson) Jin et al Journal of Translational Medicine 2010, 8:4 http://www.translational-medicine.com/content/8/1/4 Analysis of DC function and cytokine generation To measure DC cytokine production, iDC and mDCs (100,000 cells/ml) were co-incubated with 50,000 cells/ ml of adherent mouse fibroblasts transfected to express human CD40-Ligand (CD40L-LTK) in 48-well plates This cell line was kindly provided by Dr Kurlander (Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD) Before (0 hour) and after 24 hours of stimulation the supernatant was collected and the samples were analyzed by protein expression profiling The levels of 50 soluble factors were assessed on an ELISA-based platform consisting of multiplexed assays that measured up to 16 proteins per well in standard 96 well plates (Pierce Search Light Proteome Array, Boston, MA)[13] RNA preparation, amplification, and labeling for oligonucleotide microarray analysis Total RNA was extracted from the DCs using Trizol (Invitrogen, Carlsbad, CA) RNA integrity was assessed using an Agilent 2100 Bioanalyser (Agilent Technologies, Waldbronn, Germany) Total RNA (3 μg) from the DCs was amplified into anti-sense RNA (aRNA) While total RNA from PBMCs pooled from the normal donors was extracted and amplified into aRNA to serve as the reference Pooled reference and test aRNA were isolated and amplified using identical conditions and the same amplification/hybridization procedures to avoid possible interexperimental biases Both reference and test aRNA were directly labeled using ULS aRNA Fluorescent Labeling kit (Kreatech, Amsterdam, Netherlands) with Cy3 for reference and Cy5 for test samples Human oligonucleotide microarrays spanning the entire genome were printed in the Infectious Disease and Immunogenetics Section, DTM, Clinical Center, NIH using a commercial probe set containing 35,035 oligonucleotide probes, representing approximately 25,100 unique genes and 39,600 transcripts excluding control oligonucleotides (Operon Human Genome Array-Ready Oligo Set version 4.0, Huntsville, AL, USA) The design of the probe set was based on the Ensemble Human Database build (NCBI-35c), with full coverage of the NCBI human Reference sequence dataset (April 2, 2005) The microarray was composed of 48 blocks with one spot printed per probe per slide Hybridization was carried out in a water bath at 42°C for 18 to 24 hours and the arrays were then washed and scanned on a GenePix scanner Pro 4.0 (Axon, Sunnyvale, CA) with a variable photomultiplier tube to obtain optimized signal intensities with minimum (

Ngày đăng: 18/06/2014, 16:20

Từ khóa liên quan

Mục lục

  • Abstract

    • Background

    • Methods

    • Results

    • Conclusion

    • Introduction

    • Materials and methods

      • Study design

      • DC preparation, maturation, and harvest

      • Flow cytometeric analysis

      • Analysis of DC function and cytokine generation

      • RNA preparation, amplification, and labeling for oligonucleotide microarray analysis

      • miR expression analysis

      • Data processing and statistical analyses

      • miR and gene expression analysis by quantitative PCR

      • Results

        • Changes in DC antigen expression

        • Kinetics of the gene expression changes during DC maturation

        • miR expression during DC maturation

        • Proteins Produced during DC maturation

        • Mature DC function testing and cytokine detection

        • Discussion

        • Acknowledgements

Tài liệu cùng người dùng

Tài liệu liên quan