Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-33865536

ABSTRACT

The reconstructed skin micronucleus (RSMN) assay was developed in 2006, as an in vitro alternative for genotoxicity evaluation of dermally applied chemicals or products. In the years since, significant progress has been made in the optimization of the assay, including publication of a standard protocol and extensive validation. However, the diverse morphology of skin cells makes cell preparation and scoring of micronuclei (MN) tedious and subjective, thus requiring a high level of technical expertise for evaluation. This ultimately has a negative impact on throughput and the assay would benefit by the development of an automated method which could reduce scoring subjectivity while also improving the robustness of the assay by increasing the number of cells that can be scored. Imaging flow cytometry (IFC) with the ImageStream®X Mk II can capture high-resolution transmission and fluorescent imagery of cells in suspension. This proof-of-principle study describes protocol modifications that enable such automated measurement in 3D skin cells following exposure to mitomycin C and colchicine. IFC was then used for automated image capture and the Amnis® Artificial Intelligence (AAI) software permitted identification of binucleated (BN) cells with 91% precision. On average, three times as many BN cells from control samples were evaluated using IFC compared to the standard manual analysis. When IFC MNBN cells were visually scored from within the BN cell images, their frequency compared well with manual slide scoring, showing that IFC technology can be applied to the RSMN assay. This method enables faster time to result than microscope-based scoring and the initial studies presented here demonstrate its capability for the detection of statistically significant increases in MNBN frequencies. This work therefore demonstrates the feasibility of combining IFC and AAI to automate scoring for the RSMN assay and to improve its throughput and statistical robustness.


Subject(s)
Deep Learning , Flow Cytometry/methods , Image Processing, Computer-Assisted/methods , Skin/pathology , Artificial Intelligence , Automation, Laboratory/instrumentation , Automation, Laboratory/methods , False Positive Reactions , Feasibility Studies , Flow Cytometry/instrumentation , Humans , Image Processing, Computer-Assisted/instrumentation , Micronucleus Tests/instrumentation , Micronucleus Tests/methods , Mitomycin/toxicity , Models, Biological , Mutagenicity Tests/instrumentation , Mutagenicity Tests/methods , Predictive Value of Tests , Proof of Concept Study , Skin/diagnostic imaging , Skin, Artificial , Software , Tissue Scaffolds
2.
J Immunol ; 202(3): 1003-1015, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30598512

ABSTRACT

Inflammasome dysregulation is a hallmark of various inflammatory diseases. Evaluating inflammasome-associated structures (ASC specks) and caspase-1 activity by microscopy is time consuming and limited by small sample size. The current flow cytometric method, time of flight inflammasome evaluation (TOFIE), cannot visualize ASC specks or caspase-1 activity, making colocalization studies of inflammasome components and enzymatic activity impossible. We describe a rapid, high-throughput, single-cell, fluorescence-based image analysis method utilizing the Amnis ImageStreamX instrument that quantitatively and qualitatively characterizes the frequency, area, and cellular distribution of ASC specks and caspase-1 activity in mouse and human cells. Unlike TOFIE, this method differentiates between singular perinuclear specks and false positives. With our technique we also show that the presence of NLRP3 reduces the size of ASC specks, which is further reduced by the presence of active caspase-1. The capacity of our approach to simultaneously detect and quantify ASC specks and caspase-1 activity, both at the population and single-cell level, renders it the most powerful tool available for visualizing and quantifying the impact of mutations on inflammasome assembly and activity.


Subject(s)
CARD Signaling Adaptor Proteins/analysis , Caspase 1/analysis , Flow Cytometry/methods , Image Processing, Computer-Assisted/methods , Inflammasomes/metabolism , Single-Cell Analysis/methods , Fluorescence , HEK293 Cells , Humans , Macrophages , NLR Family, Pyrin Domain-Containing 3 Protein/agonists , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , THP-1 Cells
3.
Nat Immunol ; 15(4): 333-42, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24608040

ABSTRACT

Diverse cellular responses to external cues are controlled by a small number of signal-transduction pathways, but how the specificity of functional outcomes is achieved remains unclear. Here we describe a mechanism for signal integration based on the functional coupling of two distinct signaling pathways widely used in leukocytes: the ITAM pathway and the Jak-STAT pathway. Through the use of the receptor for interferon-γ (IFN-γR) and the ITAM adaptor Fcγ as an example, we found that IFN-γ modified responses of the phagocytic antibody receptor FcγRI (CD64) to specify cell-autonomous antimicrobial functions. Unexpectedly, we also found that in peritoneal macrophages, IFN-γR itself required tonic signaling from Fcγ through the kinase PI(3)K for the induction of a subset of IFN-γ-specific antimicrobial functions. Our findings may be generalizable to other ITAM and Jak-STAT signaling pathways and may help explain signal integration by those pathways.


Subject(s)
Immunoreceptor Tyrosine-Based Activation Motif/immunology , Janus Kinase 2/metabolism , Listeriosis/immunology , Macrophages/immunology , Receptor Cross-Talk/immunology , STAT1 Transcription Factor/metabolism , Animals , Bacterial Load , Cells, Cultured , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Immunoreceptor Tyrosine-Based Activation Motif/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Janus Kinase 2/genetics , Mice , Mice, Inbred Strains , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Phagocytosis/genetics , Phosphoinositide-3 Kinase Inhibitors , Protein Engineering , Receptors, IgG/genetics , Receptors, IgG/metabolism , Receptors, Interferon/metabolism , STAT1 Transcription Factor/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptional Activation/drug effects , Interferon gamma Receptor
4.
PLoS One ; 8(9): e76043, 2013.
Article in English | MEDLINE | ID: mdl-24073287

ABSTRACT

Genes are regulated at the single-cell level. Here, we performed RNA FISH of thousands of cells by flow cytometry (flow-RNA FISH) to gain insight into transcriptional variability between individual cells. These experiments utilized the murine adenocarcinoma 3134 cell line with 200 copies of the MMTV-Ras reporter integrated at a single genomic locus. The MMTV array contains approximately 800-1200 binding sites for the glucocorticoid receptor (GR) and 600 binding sites for the pioneer factor Foxa1. Hormone activation of endogenous GR by dexamethasone treatment resulted in highly variable changes in the RNA FISH intensity (25-300 pixel intensity units) and size (1.25-15 µm), indicative of probabilistic or stochastic mechanisms governing GR and cofactor activation of the MMTV promoter. Exogenous expression of the pioneer factor Foxa1 increased the FISH signal intensity and size as expected for a chromatin remodeler that enhances transcriptional competence through increased chromatin accessibility. In addition, specific analysis of Foxa1-enriched cell sub-populations showed that low and high Foxa1 levels substantially lowered the cell-to-cell variability in the FISH intensity as determined by a noise calculation termed the % coefficient of variation. These results suggest that an additional function of the pioneer factor Foxa1 may be to decrease transcriptional noise.


Subject(s)
Adenocarcinoma/genetics , Flow Cytometry , Hepatocyte Nuclear Factor 3-alpha/genetics , In Situ Hybridization, Fluorescence , Mammary Tumor Virus, Mouse/genetics , RNA/genetics , Transcriptional Activation , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Animals , Antineoplastic Agents, Hormonal/pharmacology , Chromatin Assembly and Disassembly/drug effects , DNA Probes , Dexamethasone/pharmacology , Image Processing, Computer-Assisted , Mice , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Glucocorticoid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
5.
J Leukoc Biol ; 86(3): 633-43, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19454652

ABSTRACT

Signals from stressed cells and the enteric microbiota activate macrophages and dendritic cells and mediate intestinal inflammation. HMGB1 serves as an immunogenic stimuli causing release of inflammatory cytokines by myeloid cells. Ethyl pyruvate inhibits secretion of HMGB1 and improves survival in models of endotoxemia and hemorrhagic shock. We reasoned that ethyl pyruvate may be protective in colitis, which involves similar inflammatory pathways. In IL-10(-/-) mice with established chronic colitis, ethyl pyruvate administration ameliorated colitis and reduced intestinal cytokine production. IL-10(-/-) mice demonstrated increased intestinal HMGB1 expression and decreased expression of RAGE compared with wild-type mice. Fecal HMGB1 levels were decreased in ethyl pyruvate-treated mice. Furthermore, ethyl pyruvate induced HO-1 expression in intestinal tissue. In TNBS-induced colitis, intrarectal administration of ethyl pyruvate resulted in amelioration of colitis and reduced intestinal cytokine production. In LPS-activated murine macrophages, ethyl pyruvate decreased expression of IL-12 p40 and NO production but did not affect IL-10 levels. Ethyl pyruvate did not inhibit nuclear translocation of NF-kappaB family members but attenuated NF-kappaB DNA binding. Additionally, ethyl pyruvate induced HO-1 mRNA and protein expression and HO-1 promoter activation. Moreover, ethyl pyruvate prevented nuclear-to-cytoplasmic translocation of HMGB1. In conclusion, the HMGB1/RAGE pathway has pathophysiologic and diagnostic significance in experimental colitis. Ethyl pyruvate and other strategies to inhibit HMGB1 release and function represent promising interventions in chronic inflammatory diseases.


Subject(s)
Colitis/metabolism , HMGB1 Protein/metabolism , Pyruvates/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/secondary , Animals , Annexin A5/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colitis/chemically induced , Colitis/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Colonic Neoplasms/secondary , Cytokines/antagonists & inhibitors , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Fluorescein-5-isothiocyanate/metabolism , In Situ Nick-End Labeling , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukin-12 Subunit p40/metabolism , Intestinal Mucosa/metabolism , Liver Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Neoplasm Metastasis , Neoplasm Transplantation , Nitric Oxide/metabolism , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Pyruvates/pharmacology , Random Allocation , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Transfection
6.
J Immunother ; 30(6): 596-606, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17667523

ABSTRACT

We have recently demonstrated that cytolysis of human melanoma cells by immune effectors (both NK and T cells) is associated with release of the nuclear chromatin protein, high mobility group box I (HMGB1). Extracellular HMGB1 mediates a number of important functions including endothelial cell activation, stromagenesis, recruitment and activation of innate immune cells, and also dendritic cell maturation that, in the setting of cancer, lead to a chronic inflammatory response. This reparative inflammatory response promotes tumor cell survival, expansion, and metastases. Release of HMGB1 after chemotherapy-induced cytotoxicity has not been well characterized. We measured the release of HMGB1 after chemotherapy or immune cytolysis and demonstrated that this did not correlate with conventional markers of apoptosis and necrosis in several human colorectal, pancreatic, and melanoma tumor cell lines. Rather, we observed that tumor cells incubated with the platinating agent oxaliplatin, retained HMGB1 within the nucleus for significantly longer periods than other agents used at comparable cytotoxic concentrations or even with potent cytolytic cells. Thus, release of HMGB1 from dying tumor cells treated with chemotherapy or cells with lymphokine activated killer cell activity is not dependent solely on the mode of cell death. Sequestration of the damage associated molecular pattern molecule, HMGB1, may play a role in the clinical efficacy of platinating agents and suggests this as a superior agent for coupling with immunotherapeutic strategies, possibly enhancing their effectiveness.


Subject(s)
Antineoplastic Agents/pharmacology , HMGB1 Protein/metabolism , Neoplasms/metabolism , Neoplasms/therapy , Organoplatinum Compounds/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Line, Tumor , Cell Nucleus/metabolism , Colonic Neoplasms , Combined Modality Therapy , Humans , Immunotherapy , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Lymphokine-Activated/metabolism , Melanoma , Melphalan/pharmacology , Microscopy, Confocal , Necrosis , Neoplasms/drug therapy , Neoplasms/pathology , Oxaliplatin , Paclitaxel/pharmacology , Pancreatic Neoplasms
7.
J Leukoc Biol ; 81(1): 75-83, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16968820

ABSTRACT

High mobility group box 1 (HMGB1) is one of the recently defined damage-associated molecular pattern molecules, passively released from necrotic cells and secreted by activated macrophage/monocytes. Whether cytolytic cells induce HMGB1 release from tumor cells is not known. We developed a highly sensitive method for detecting intracellular HMGB1 in tumor cells, allowing analysis of the type of cell death and in particular, necrosis. We induced melanoma cell death with cytolytic lymphokine-activated killing (LAK) cells, tumor-specific cytolytic T lymphocytes, TRAIL, or granzyme B delivery and assessed intracellular HMGB1 retention or release to investigate the mechanism of HMGB1 release by cytolytic cells. HMGB1 release from melanoma cells (451Lu, WM9) was detected within 4 h and 24 h following incubation with IL-2-activated PBMC (LAK activity). HLA-A2 and MART1 or gp100-specific cytolytic T lymphocytes induced HMGB1 release from HLA-A2-positive and MART1-positive melanoma cells (FEM X) or T2 cell-loaded, gp100-specific peptides. TRAIL treatment, however, induced HMGB1 release, and it is interesting that this extrinsic pathway-mediated cell death was blocked with the pancaspase inhibitor N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone. Conversely, granzyme B delivery did not induce HMGB1 release. HMGB1, along with other intracellular factors released from tumor cells induced by cytolysis, may be important components of the disordered tumor microenvironment. This has important implications for the immunotherapy of patients with cancer. Specifically, HMGB1 may promote healing or immune reactivity, depending on the nature of the local inflammatory response and the presence (or absence) of immune effectors.


Subject(s)
HMGB1 Protein/metabolism , Melanoma/metabolism , T-Lymphocytes/physiology , Active Transport, Cell Nucleus , Amino Acid Chloromethyl Ketones/pharmacology , Cell Death , Cell Line, Tumor , Cell Nucleus/metabolism , Cells, Cultured , Cytotoxicity Tests, Immunologic , Flow Cytometry , Granzymes/pharmacology , Humans , Killer Cells, Lymphokine-Activated/physiology , TNF-Related Apoptosis-Inducing Ligand/pharmacology
8.
J Immunol ; 176(12): 7154-8, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16751357

ABSTRACT

High mobility group box 1 (HMGB1) is a NF released extracellularly as a late mediator of lethality in sepsis and as an early mediator of inflammation following injury. Here we demonstrate that in contrast to the proinflammatory role of HMGB1, preconditioning with HMGB1 results in protection following hepatic ischemia/reperfusion (I/R). Pretreatment of mice with HMGB1 significantly decreased liver damage after I/R. The protection observed in mice pretreated with HMGB1 was associated with a higher expression of IL-1R-associated kinase-M, a negative regulator of TLR4 signaling, compared with controls. We thus explored the possibility that HMGB1 preconditioning was mediated through TLR4 activation. HMGB1 preconditioning failed to provide protection in TLR4 mutant (C3H/HeJ) mice, but successfully reduced damage in TLR4 wild-type (C3H/HeOuj) mice. Our studies demonstrate that in contrast to the role of HMGB1 as an early mediator of inflammation and organ damage in hepatic I/R, HMGB1 preconditioning can be protective.


Subject(s)
HMGB1 Protein/administration & dosage , Liver/blood supply , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Transplantation Conditioning/methods , Amino Acid Sequence , Animals , HMGB1 Protein/genetics , HeLa Cells , Humans , Injections, Intravenous , Interleukin-1 Receptor-Associated Kinases , Intracellular Signaling Peptides and Proteins , Lipopolysaccharides/administration & dosage , Liver/pathology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Sequence Data , Protein Serine-Threonine Kinases/biosynthesis , Reperfusion Injury/enzymology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/physiology , Up-Regulation/physiology
9.
J Heart Lung Transplant ; 25(3): 310-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16507425

ABSTRACT

BACKGROUND: Administration of immature dendritic cells (DC) prolongs but does not result in indefinite allograft survival. We attempted to achieve this goal by adding a sub-therapeutic dose of immunosuppression. METHODS: DC propagated from B10 (H-2(b)) mouse bone marrow (BM) were transfected with nuclear factor-kappaB (NF-kappaB)-binding-site-specific oligodeoxyribonucleotide (ODN). The allostimulatory activity of transfected and normal DC were examined in mixed-lymphocyte reaction (MLR) and cytotoxic T-lymphocyte (CTL) assays in vitro, and their influence on allograft survival by systemic administration of DC in vivo. RESULTS: Transfection of DC with NF-kappaB ODN resulted in complete abrogation of NF-kappaB activity and inhibition of co-stimulation. Allogeneic (C3H, H-2(k)) T cells stimulated by ODN DC demonstrated impairment in MLR and CTL activity. Administration of ODN DC significantly prolonged B10 allograft survival. In contrast to cyclosporine, which failed to enhance the effect of ODN DC, a combination of ODN DC with sirolimus at 6 mg/kg/day for 6 days achieved long-term survival in all allografts. This was associated with low CTL activity of either graft-infiltrating cells or splenic T cells and increased TUNEL-positive cells in T-cell areas of recipient mesenteric lymph nodes. Analysis of transcription factor nuclear translocation with Cellomics indicated that stimulation with ODN DC showed inhibited T-cell nuclear translocation of signal transducer and activator of transcription (Stat)1 and Stat3, extracellular signal-related kinase (ERK) and activating transcription factor (ATF)-2, but not NF-kappaB and P38, compared with mature DC. The selective inhibition was enhanced by sirolimus, but not cyclosporine. CONCLUSIONS: Sirolimus enhances immature DC tolerogenicity by induction of T-cell apoptosis, and promotes immature DC-induced inhibition of Stat1, ERK and ATF-2 activation.


Subject(s)
Dendritic Cells/transplantation , Graft Survival/drug effects , Graft Survival/physiology , Heart Transplantation , Immunosuppressive Agents/administration & dosage , Sirolimus/administration & dosage , T-Lymphocytes, Cytotoxic/physiology , Animals , Apoptosis/drug effects , Dendritic Cells/drug effects , Flow Cytometry , Lymphocyte Culture Test, Mixed , Mice , NF-kappa B/drug effects , NF-kappa B/metabolism , Oligodeoxyribonucleotides/pharmacology , T-Lymphocytes, Cytotoxic/drug effects , Transcription Factors/drug effects , Transfection
10.
Drug Discov Today ; Suppl: 19-30, 2005.
Article in English | MEDLINE | ID: mdl-23570163

ABSTRACT

Rapid assessment of immune or stem cells, which are now widely applied in the clinical setting of cancer treatment, is necessary to speed their development and to determine their quality. We have evaluated immature dendritic cells (iDC) by semiautomated imaging cytometry which provides detailed assessment at a single cell level. Nuclear translocation of NF-kappaB was studied by imaging analysis as well as electrophoretic mobility shift assay with an excellent correlation (r = 0.981) over a broad range of lipopolysaccharide (LPS) concentrations. Imaging analysis was time saving (5 h vs. 3 days), and required 30- to 100-fold less cells per analysis. Single cell information revealed remarkable heterogeneity between individual iDC and permitted detection of responses to 40 pg/ml of LPS. In IL-1beta/IFNgamma activated iDC, STAT1 responses preceded NF-kappaB responses, and the expression of both was strongly correlated in individual cells (p < 0.001). IFNgamma amplified IL-1-induced NF-kappaB responses. NF-kappaB responses to IL-1beta, CD40L, and LPS were donor-dependent (n = 7), correlated with the quality of iDC preparations (p = 0.002), and IL-12 p70 production (p = 0.010). NF-kappaB measurements in iDC within mixed cell cultures (iDC, NK, K562) demonstrated that these strategies are applicable for analyses of complex cell-cell interactions. Imaging analysis is a method that could be valuable for quality control of cell therapy preparations.


Subject(s)
Dendritic Cells/metabolism , NF-kappa B/metabolism , STAT1 Transcription Factor/metabolism , Single-Cell Analysis , Active Transport, Cell Nucleus , Electrophoretic Mobility Shift Assay , Flow Cytometry , Humans , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Protein Transport
11.
Mol Immunol ; 42(4): 433-44, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15607795

ABSTRACT

Substantial attention has been paid to the role of the toll-like receptor (TLR) ligands of late and their role in regulating the innate immune response. They serve as exogenous danger signals important in informing and driving the distal adaptive immune response to pathogens. Less clear has been the role of the nominal endogenous danger signals released and recognized in stressed cells following genotoxic or metabolic stress as occurs in progressively growing tumors. HMGB1 (high-mobility group B1) is a nuclear protein well characterized for its ability to modify DNA access to transcriptional proteins that is released from necrotic cells as well as secreted through the endosomal route from hematopoietic cells, serving as a late mediator of sepsis. It interacts with high-affinity RAGE (receptor for advanced glycation end products) and TLR2 receptors. Here we show that HMGB1 enhances interferon gamma release from macrophage (but not dendritic cell)-stimulated NK cells. This is effective only when coupled with other pro-inflammatory cytokines particularly with IL-2 in combination with IL-1 or IL-12. We have used this information to suggest that HMGB1, which also promotes epithelial migration and proliferation, drives repair in the absence or inhibition of other factors but enhances inflammation in their presence. The implications for tumorigenesis and tumor progression are quite important as they may be for other states of chronic inflammation.


Subject(s)
Cell Communication/immunology , HMGB1 Protein/physiology , Interferon-gamma/biosynthesis , Killer Cells, Natural/immunology , Macrophages/immunology , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/immunology , HMGB1 Protein/pharmacology , Humans , Immunity, Innate , Interferon-gamma/immunology , Interleukins/biosynthesis , Interleukins/immunology , Macrophages/drug effects , Membrane Glycoproteins/immunology , Membrane Glycoproteins/physiology , Monocytes/immunology , Receptors, Cell Surface/immunology , Receptors, Cell Surface/physiology , Signal Transduction , Toll-Like Receptor 2 , Toll-Like Receptors
12.
J Immunol Methods ; 294(1-2): 123-34, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15604022

ABSTRACT

Rapid assessment of immune or stem cells, which are now widely applied in the clinical setting of cancer treatment, is necessary to speed their development and to determine their quality. We have evaluated immature dendritic cells (iDC) by semiautomated imaging cytometry which provides detailed assessment at a single cell level. Nuclear translocation of NF-kappaB was studied by imaging analysis as well as electrophoretic mobility shift assay with an excellent correlation (r=0.981) over a broad range of lipopolysaccharide (LPS) concentrations. Imaging analysis was time saving (5 h vs. 3 days), and required 30- to 100-fold less cells per analysis. Single cell information revealed remarkable heterogeneity between individual iDC and permitted detection of responses to 40 pg/ml of LPS. In IL-1beta/IFNgamma activated iDC, STAT1 responses preceded NF-kappaB responses, and the expression of both was strongly correlated in individual cells (p<0.001). IFNgamma amplified IL-1-induced NF-kappaB responses. NF-kappaB responses to IL-1beta, CD40L, and LPS were donor-dependent (n=7), correlated with the quality of iDC preparations (p=0.002), and IL-12 p70 production (p=0.010). NF-kappaB measurements in iDC within mixed cell cultures (iDC, NK, K562) demonstrated that these strategies are applicable for analyses of complex cell-cell interactions. Imaging analysis is a method that could be valuable for quality control of cell therapy preparations.


Subject(s)
Cell Nucleus/physiology , DNA-Binding Proteins/metabolism , Dendritic Cells/physiology , Microscopy, Fluorescence , NF-kappa B/metabolism , Trans-Activators/metabolism , Antineoplastic Agents/pharmacology , CD40 Ligand/metabolism , Cell Communication/physiology , Cell Separation , Cell- and Tissue-Based Therapy , Dendritic Cells/cytology , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Interleukin-12/biosynthesis , K562 Cells , Killer Cells, Natural/cytology , Killer Cells, Natural/physiology , Lipopolysaccharides/pharmacology , Protein Transport/drug effects , Protein Transport/physiology , Quality Control , STAT1 Transcription Factor , Signal Transduction/drug effects , Signal Transduction/physiology
13.
Discov Med ; 4(24): 448-56, 2004 Dec.
Article in English | MEDLINE | ID: mdl-20704947

ABSTRACT

Extract: We all shudder about untimely deaths or those that we were not prepared for. As such we perceive such "unscheduled" deaths as dangerous. Similarly, apoptotic death (literally falling leaves) or the programmed cell death of cells in multicellular organisms ranging from slime mold and simple worms through to mammals, has a level of tidiness and well-orchestrated activities with literally hundreds if not thousands of gene products employed with either the primary or secondary purpose of coordinating the orderly death of cells throughout life. During inflammation of any sort, driven by tissue damage or injury or infection by pathogens (virus, bacteria, and parasites), apoptotic death similarly serves to quickly rid the host of damaged cells, promote removal and digestion of the infected cell, and prepare the way for tissue remodeling and repair. When this goes awry, for example during periods of chronic inflammation, tissues are subjected to the contrasting needs of driving apoptotic death whilst maintaining the barrier function of the epithelia (such as skin cells) as well as the selective permeability of mucosal sites (i.e., areas where mucus is secreted to protect the cells from their surroundings, such as gut cells protecting themselves from the gastric acids). Prudently, they need to limit and husband local resources sufficiently for the maintenance of tissue integrity and renewal. It is our provocative and novel contention that cancer in adults (and not children) most often arises in a setting of chronic inflammation and disordered cell death rather than one associated primarily with disordered cell growth as it is popularly imagined by scientists, clinicians, and the general public.

14.
Curr Opin Investig Drugs ; 4(12): 1405-9, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14763124

ABSTRACT

High mobility group B1 (HMGB1) and its counter-receptor, receptor for advanced glycation end products (RAGE), represent suitable targets for investigation, integrating many aspects of modern biology, particularly that associated with chronic diseases involving inflammation, dysregulated cell death and cancer. Also known as amphoterin, HMGB1 was initially identified over 25 years ago as a transcriptional regulatory molecule causing DNA bending, and facilitating binding of several transcriptional complexes, in particular members of the nuclear hormone receptor family. Although loosely bound to chromatin, it is released from necrotic cells but not apoptotic cells and is actively secreted by activated macrophages in a partially tumor necrosis factor-dependent manner. It serves as a late mediator of septic death present within the serum and inflammatory sites of patients with arthritis, correlating with the inflammatory response, to signal tissue injury, causes sickness behavior, and acts as an endogenous pyrogen. Although known to interact with RAGE on endothelial cells causing activation and leukocyte recruitment, RAGE itself has most recently been shown to serve as a counter-receptor for leukocyte integrins, suggesting that signaling through this molecule is potentially important for cell adhesion and clustering as well as recruitment of inflammatory cells. Targeting the HMGB1 ligand or its receptor represents an important potential application in cancer therapeutics, given its widespread overexpression, as well as that of its receptor in virtually every tumor type carefully examined. This, coupled with its ability to accelerate tumor growth in immunodeficient murine models, suggests that it is a possible therapeutic target in patients with cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Delivery Systems/methods , HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/physiology , Neoplasms/drug therapy , Neoplasms/pathology , Receptors, Immunologic/metabolism , Animals , Cell Death/drug effects , Glycation End Products, Advanced , HMGB1 Protein/metabolism , Humans , Neoplasms/enzymology , Neoplasms/metabolism , Receptor for Advanced Glycation End Products
15.
J Immunol ; 168(4): 1618-26, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11823489

ABSTRACT

Screening expressed sequence tag databases for endothelial-specific homologs to human junctional adhesion molecule (JAM) and A33-Ag, we identified a protein of 298 aa that represents the recently described vascular endothelial-JAM (VE-JAM)/JAM 2. We confirmed VE-JAM/JAM 2 expression to be restricted to the high endothelial venule of tonsil and lymph nodes, and we further expanded the localization to the endothelium of arterioles in and around inflammatory and tumor foci. In our functional characterizations of VE-JAM/JAM 2, we discovered that it can function as an adhesive ligand for the T cell line J45 and can interact with GM-CSF/IL-4-derived peripheral blood dendritic cells, circulating CD56(+) NK cells, circulating CD56(+)CD3(+) NK/T cells, and circulating CD56(+)CD3(+)CD8(+) cytolytic T cells. In the course of our studies, we also isolated and characterized the functional VE-JAM/JAM 2 receptor, which, upon cloning, turned out to be a submitted sequence representing JAM 3 (accession number NP 113658). With these understandings, we have characterized a protein-interacting pair that can be important in the role of T, NK, and dendritic cell trafficking and inflammation.


Subject(s)
Cell Adhesion Molecules/metabolism , Dendritic Cells/immunology , Immunoglobulins/metabolism , Killer Cells, Natural/immunology , Membrane Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Animals , CHO Cells , Cell Adhesion Molecules/genetics , Cell Line , Cloning, Molecular , Cricetinae , Endothelium, Vascular/metabolism , Humans , Immunoglobulins/genetics , In Situ Hybridization , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Leukocytes/immunology , Membrane Proteins/genetics , Molecular Sequence Data , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , RNA, Messenger/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...