Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Arch Toxicol ; 98(9): 2985-2998, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38806719

ABSTRACT

The development of inhaled drugs for respiratory diseases is frequently impacted by lung pathology in non-clinical safety studies. To enable design of novel candidate drugs with the right safety profile, predictive in vitro lung toxicity assays are required that can be applied during drug discovery for early hazard identification and mitigation. Here, we describe a novel high-content imaging-based screening assay that allows for quantification of the tight junction protein occludin in A549 cells, as a model for lung epithelial barrier integrity. We assessed a set of compounds with a known lung safety profile, defined by clinical safety or non-clinical in vivo toxicology data, and were able to correctly identify 9 of 10 compounds with a respiratory safety risk and 9 of 9 compounds without a respiratory safety risk (90% sensitivity, 100% specificity). The assay was sensitive at relevant compound concentrations to influence medicinal chemistry optimization programs and, with an accessible cell model in a 96-well plate format, short protocol and application of automated imaging analysis algorithms, this assay can be readily integrated in routine discovery safety screening to identify and mitigate respiratory toxicity early during drug discovery. Interestingly, when we applied physiologically-based pharmacokinetic (PBPK) modelling to predict epithelial lining fluid exposures of the respiratory tract after inhalation, we found a robust correlation between in vitro occludin assay data and lung pathology in vivo, suggesting the assay can inform translational risk assessment for inhaled small molecules.


Subject(s)
Lung , Occludin , Humans , Lung/drug effects , Lung/pathology , A549 Cells , Occludin/metabolism , Toxicity Tests/methods , High-Throughput Screening Assays/methods , Administration, Inhalation , Drug Discovery/methods
2.
Mol Cancer ; 16(1): 73, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28372546

ABSTRACT

BACKGROUND: The role of cancer-associated fibroblasts (CAFs) during tumour progression is obscured by the inherently complex, heterotypic nature of fibroblast cells and behaviours in various subtypes of malignancies. Therefore, we sought to identify distinct fibroblast subpopulations at the single-cell level. METHODS: Using single-cell quantitative PCR as a powerful tool to study heterogeneity and rare cell events, in a high-throughput manner a panel of gene targets are run simultaneously on transcripts isolated from single cells obtained by fluorescence-activated cell sort. Assessment of cells with stem-like characteristics was attained by anchorage-independent, anoikis-resistant culture. RESULTS: Single-cell analysis of fibroblasts and their tumour-activated counterparts demonstrated molecularly distinct cell types defined by differential expression of characteristic mesenchymal and fibroblast activation markers. Identified subpopulations presented overlapping gene expression patterns indicating transitional molecular states during fibroblast differentiation. Using single-cell resolution data we generated a molecular differentiation model which enabled the classification of patient-derived fibroblasts, validating our modelling approach. Remarkably, a subset of fibroblasts displayed expression of pluripotency markers, which was enriched for in non-adherent conditions. Yet the ability to form single-cell derived spheres was generally reduced in CAFs and upon fibroblast activation through TGFß1 ligand and cancer cell-secreted factors. Hence, our data imply the existence of putative stem/progenitor cells as a physiological feature of undifferentiated fibroblasts. CONCLUSIONS: Within this comprehensive study we have identified distinct and intersecting molecular profiles defining fibroblast activation states and propose that underlying cellular heterogeneity, fibroblasts are hierarchically organized. Understanding the molecular make-up of cellular organization and differentiation routes will facilitate the discovery of more specific markers for stromal subtypes and targets for anti-stromal therapies.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Anoikis/genetics , Biomarkers, Tumor , Cell Line , Cluster Analysis , Female , Gene Expression Profiling , Humans , Single-Cell Analysis , Tumor Microenvironment
3.
Antioxidants (Basel) ; 13(8)2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39199170

ABSTRACT

Oxidative stress in the human lung is caused by both internal (e.g., inflammation) and external stressors (smoking, pollution, and infection) to drive pathology in a number of lung diseases. Cellular damage caused by oxidative damage is reversed by several pathways, one of which is the antioxidant response. This response is regulated by the transcriptional factor NRF2, which has the ability to regulate the transcription of more than 250 genes. In disease, this balance is overwhelmed, and the cells are unable to return to homeostasis. Several pharmacological approaches aim to improve the antioxidant capacity by inhibiting the interaction of NRF2 with its key cytosolic inhibitor, KEAP1. Here, we evaluate an alternative approach by overexpressing NRF2 from chemically modified RNAs (cmRNAs). Our results demonstrate successful expression of functional NRF2 protein in human cell lines and primary cells. We establish a kinetic transcriptomic profile to compare antioxidant response gene expression after treatment of primary human bronchial epithelial cells with either KEAP1 inhibitors or cmRNAs. The key gene signature is then applied to primary human lung fibroblasts and alveolar macrophages to uncover transcriptional preferences in each cell system. This study provides a foundation for the understanding of NRF2 dynamics in the human lung and provides initial evidence of alternative ways for pharmacological interference.

4.
Exp Cell Res ; 315(10): 1724-33, 2009 Jun 10.
Article in English | MEDLINE | ID: mdl-19331815

ABSTRACT

The intracellular signaling molecule mammalian target of rapamycin (mTOR) is essential for cell growth and proliferation. It is involved in mouse embryogenesis, murine trophoblast outgrowth and linked to tumor cell invasiveness. In order to assess the role of mTOR in human trophoblast invasion we analyzed the in vitro invasiveness of HTR-8/SVneo immortalized first-trimester trophoblast cells in conjunction with enzyme secretion upon mTOR inhibition and knockdown of mTOR protein expression. Additionally, we also tested the capability of mTOR to trigger signal transducer and activator of transcription (STAT)-3 by its phosphorylation status. Rapamycin inhibited mTOR kinase activity as demonstrated with a lower phosphorylation level of the mTOR substrate p70 S6 kinase (S6K). With the use of rapamycin and siRNA-mediated mTOR knockdown we could show that cell proliferation, invasion and secretion of matrix-metalloproteinases (MMP)-2 and -9, urokinase-like plasminogen activator (uPA) and its major physiological uPA inhibitor (PAI)-1 were inhibited. While tyrosine phosphorylation of STAT3 was unaffected by mTOR inhibition and knockdown, serine phosphorylation was diminished. We conclude that mTOR signaling is one major mechanism in a tightly regulated network of intracellular signal pathways including the JAK/STAT system to regulate invasion in human trophoblast cells by secretion of enzymes that remodel the extra-cellular matrix (ECM) such as MMP-2, -9, uPA and PAI-1. Dysregulation of mTOR may contribute to pregnancy-related pathologies caused through impaired trophoblast invasion.


Subject(s)
Cell Movement , Extracellular Matrix/enzymology , Phosphoserine/metabolism , Protein Kinases/metabolism , STAT3 Transcription Factor/metabolism , Trophoblasts/cytology , Trophoblasts/enzymology , Blotting, Western , Cell Extracts , Cell Proliferation , Colorimetry , Culture Media, Conditioned , Female , Humans , Models, Biological , Phosphorylation , Plasminogen Activator Inhibitor 1/metabolism , Pregnancy , TOR Serine-Threonine Kinases
5.
Chem Immunol Allergy ; 88: 181-199, 2005.
Article in English | MEDLINE | ID: mdl-16129946

ABSTRACT

During the first trimester of pregnancy, well-differentiated primary cells of the placenta known as trophoblast cells grow in an invasive and destructive fashion similar to malignancies, but limited in space and time. The comparison of trophoblast cells with their malignant counterpart, human choriocarcinoma cells, offers an attractive model to understand the origin or development of malignant growth. Several cytokines and growth factors are known to influence trophoblast migration (e.g. EGF, IGF-2, HGF), proliferation (e.g. leptin, HGF, GM-CSF) and/or invasion (e.g. leukemia inhibitory factor, LIF), each factor utilizing at least one pathway for intracellular signaling in the trophoblast. Two pathways that are crossed especially often mediate the signals of these factors and are simultaneously well established in terms of tumor invasion: the Janus kinase-signal transducers and activators of transcription (Jak-Stat) and receptor-associated tyrosine kinase-mitogen-activated protein kinase (RTK-MAPK) pathways. These two pathways are detrimental for reproduction in general, and in part for placenta development, as a series of knockout experiments demonstrate. Aspects of each pathway are also implicated to be involved in trophoblast invasion, e.g. STAT3 is constitutively activated in invasive first trimester trophoblast cells, and activated ERK is detectable in intermediate trophoblast cells, an invasive phenotype. Interaction at several intersection points between the pathways has been described in several cell systems so that the same would seem to be possible in trophoblast cells. In this review, some of the possible areas of interaction are alluded to.


Subject(s)
Cell Movement , Signal Transduction , Trophoblasts/cytology , Trophoblasts/metabolism , Animals , Humans , Neoplasms/metabolism , Neoplasms/pathology , Protein-Tyrosine Kinases/metabolism , ras Proteins/metabolism
6.
Chem Immunol Allergy ; 89: 26-35, 2005.
Article in English | MEDLINE | ID: mdl-16129950

ABSTRACT

Pregnancy is accompanied by a Th2-prone immune modulation, which is a major puzzle piece among maternofetal tolerance-promoting factors. A large number of cytokines is physiologically or pathologically present in the decidua and is potentially able to act on lymphocytes and NK cells, which express a variety of respective receptors. Intracellular signals from these receptors are to a major part transduced via the Janus kinases (JAK) and signal transducers and activators of a transcription (STAT) system, which consists of at least 4 different kinases and 7 STATs plus several subtypes and splicing variants. A network of suppressors of cytokine signaling (SOCS) controls their balance. The interactions of all these intracellular factors and cross-linking with further signaling systems seem to be crucial for the maintenance of a maternal cytokine profile which promotes the tolerance of the fetus.


Subject(s)
Lymphocytes/immunology , Lymphocytes/metabolism , Maternal-Fetal Exchange/immunology , Animals , Cytokines/metabolism , Decidua/immunology , Decidua/metabolism , Female , Growth Substances/metabolism , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Pregnancy , Protein-Tyrosine Kinases/immunology , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Trans-Activators/immunology
7.
Cancer Res ; 75(7): 1457-69, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25833830

ABSTRACT

One third of the patients with estrogen receptor α (ERα)-positive breast cancer who are treated with the antiestrogen tamoxifen will either not respond to initial therapy or will develop drug resistance. Endocrine response involves crosstalk between ERα and TGFß signaling, such that tamoxifen nonresponsiveness or resistance in breast cancer might involve aberrant TGFß signaling. In this study, we analyzed TGFß receptor type 2 (TGFBR2) expression and correlated it with ERα status and phosphorylation in a cohort of 564 patients who had been randomized to tamoxifen or no-adjuvant treatment for invasive breast carcinoma. We also evaluated an additional four independent genetic datasets in invasive breast cancer. In all the cohorts we analyzed, we documented an association of low TGFBR2 protein and mRNA expression with tamoxifen resistance. Functional investigations confirmed that cell cycle or apoptosis responses to estrogen or tamoxifen in ERα-positive breast cancer cells were impaired by TGFBR2 silencing, as was ERα phosphorylation, tamoxifen-induced transcriptional activation of TGFß, and upregulation of the multidrug resistance protein ABCG2. Acquisition of low TGFBR2 expression as a contributing factor to endocrine resistance was validated prospectively in a tamoxifen-resistant cell line generated by long-term drug treatment. Collectively, our results established a central contribution of TGFß signaling in endocrine resistance in breast cancer and offered evidence that TGFBR2 can serve as an independent biomarker to predict treatment outcomes in ERα-positive forms of this disease.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Estrogens/physiology , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Tamoxifen/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Antineoplastic Agents, Hormonal/therapeutic use , Apoptosis , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/mortality , Cell Proliferation , Disease-Free Survival , Drug Resistance, Neoplasm , Female , Follow-Up Studies , Gene Expression , Gene Knockdown Techniques , HEK293 Cells , Humans , Kaplan-Meier Estimate , MCF-7 Cells , Multivariate Analysis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Proportional Hazards Models , Protein Serine-Threonine Kinases/metabolism , Receptor Cross-Talk , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Tamoxifen/therapeutic use
8.
PLoS One ; 7(9): e45669, 2012.
Article in English | MEDLINE | ID: mdl-23029174

ABSTRACT

PURPOSE: The aim of this study was to evaluate ERK phosphorylation as a stromal biomarker for breast cancer prognosis and tamoxifen treatment prediction within a randomized tamoxifen trial. PATIENTS AND METHODS: Tissue microarrays of two breast cancer cohorts including in total 743 invasive breast cancer samples were analyzed for ERK phosphorylation (pERK) and smooth muscle actin-alpha expression (SMAα) in cancer-associated fibroblasts (CAFs) and links to clinico-pathological data and treatment-predictive values were delineated. RESULTS: By analyzing a unique randomized tamoxifen trial including breast cancer patients receiving no adjuvant treatment we show for the first time that patients low in ERK phosphorylation in CAFs did not respond to tamoxifen treatment despite having estrogen-receptor alpha (ERα-positive tumors compared to patients with high pERK levels in CAFs (P = 0.015, multivariate Cox regression interaction analysis). In both clinical materials we further show a significant association between pERK and SMAα, a characteristic marker for activated fibroblasts. SMAα expression however was not linked to treatment-predictive information but instead had prognostic qualities. CONCLUSION: The data suggests that the presence of a subpopulation of CAFs, defined by minimal activated ERK signaling, is linked to an impaired tamoxifen response. Thus, this report illustrates the importance of the stroma for monitoring treatment effects in pre-menopausal breast cancer.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Premenopause , Tamoxifen/therapeutic use , Actins/metabolism , Biomarkers, Tumor/metabolism , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Cohort Studies , Estrogen Receptor alpha/metabolism , Female , Fibroblasts/enzymology , Humans , Phosphorylation , Tissue Array Analysis
9.
PLoS One ; 7(4): e36051, 2012.
Article in English | MEDLINE | ID: mdl-22563438

ABSTRACT

BACKGROUND: MicroRNAs are small non-coding RNAs involved in the regulation of gene expression on a posttranscriptional level. These regulatory RNAs have been implicated in numerous cellular processes and are further deregulated in different cancer types, including breast cancer. MiR-92a is part of the miR-17∼92 cluster, which was first reported to be linked to tumourigenesis. However, little is known about the expression of miR-92a in breast cancer and potential associations to tumour properties. The expression of miR-92a was therefore characterized in 144 invasive breast cancer samples using in situ hybridization and related to clinico-pathological data as well as to selected key properties of the tumour stroma, including the presence of macrophages (CD68) and cancer activated fibroblasts (alpha-SMA). METHODOLOGY/PRINCIPAL FINDINGS: To measure miR-92a levels, an in situ hybridisation protocol was developed and validated using cell lines and miR-92a inhibitors. The expression in the tumour samples was objectively evaluated using digital image analysis program subtracting background activities. We found that the miR-92a expression varied between tumours and was inversely correlated to tumour grade (r = -0.276, p = 0.003) and recurrence-free survival (p = 0.008) and provided independent prognostic information in multivariate Cox analysis (HR: 0.375, CI: 0.145-0.972, p = 0.043). MiR-92a was moreover inversely correlated to the number of infiltrating macrophages in the tumour stroma (r = -0.357, p<0.001), and downregulation of miR-92a promoted cell migration (p<0.01). CONCLUSIONS/SIGNIFICANCE: This study demonstrates that downregulation of miR-92a in breast cancer is linked to key epithelial and stromal properties as well as clinical outcome.


Subject(s)
Breast Neoplasms/metabolism , Macrophages/physiology , MicroRNAs/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Disease-Free Survival , Down-Regulation , Female , Humans , In Situ Hybridization , Neoplasm Staging , Prognosis , Software
10.
Am J Reprod Immunol ; 66(4): 329-35, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21385272

ABSTRACT

OBJECTIVES In previous studies, we have shown that sHLA-G reduces cytotoxicity of decidual NK cells, which was dependent upon reduction in signal transducer and activator of transcription 3 (STAT3) and perforin. In this study, we aimed to confirm the role of STAT3 for induction of cytotoxicity and to analyze the regulative role of its antagonist suppressor of cytokine signaling 3 (SOCS3). Furthermore, the influence of both factors on cytokine expression should be analyzed. METHODS All experiments were performed on NK-92 cells. STAT3 and SOCS3 have been silenced using two different small interfering RNA sequences each. Silencing efficiency and STAT3 tyrosine phosphorylation have been analyzed by Western blotting. Cytotoxicity to K562 target cells has been assessed by flow cytometry. Expression of IFN-γ, IL-1ß, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, TNF-α, and TNF-ß has been measured using cytometric bead arrays for flow cytometry. RESULTS STAT3 and SOCS3 have been successfully silenced. STAT3 silencing reduced cytotoxicity. SOCS3 silencing induced increase in STAT3 tyrosine phosphorylation and cytotoxicity. STAT3 silencing reduced IL-10 expression significantly, while SOCS3 silencing induced, also significantly, the opposite effect. The other cytokines were expressed at very low concentration or not constantly affected. CONCLUSION STAT3 and SOCS3 are involved in regulation of NK cell cytotoxicity and IL-10 expression.


Subject(s)
Decidua/immunology , Interleukin-10/biosynthesis , Killer Cells, Natural/immunology , STAT3 Transcription Factor/immunology , Signal Transduction/immunology , Suppressor of Cytokine Signaling Proteins/immunology , Blotting, Western , Cell Line , Cytotoxicity Tests, Immunologic , Decidua/cytology , Decidua/metabolism , Female , Flow Cytometry , Gene Silencing/drug effects , Gene Silencing/immunology , Humans , Interleukin-10/immunology , K562 Cells , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Phosphorylation/immunology , Pregnancy , RNA, Small Interfering/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/antagonists & inhibitors , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Tyrosine/metabolism
11.
Oncoscience ; 2(10): 835-6, 2015.
Article in English | MEDLINE | ID: mdl-26682269
12.
J Biol Chem ; 282(25): 18542-18551, 2007 Jun 22.
Article in English | MEDLINE | ID: mdl-17470430

ABSTRACT

The small GTPase Rheb is a positive upstream regulator of the target of rapamycin (TOR) complex 1 in mammalian cells and can bind directly to TOR complex 1. To identify the regions of the Rheb surface most critical for signaling to TOR complex 1, we created a set of 26 mutants wherein clusters of 1-5 putative solvent-exposed residues were changed to alanine, ultimately changing 65 residues distributed over the entire Rheb surface. The signaling function of these mutants was assessed by their ability, in comparison to wild type Rheb, to restore the phosphorylation of S6K1(Thr389) when expressed transiently in amino acid-deprived 293T cells. The major finding is that two mutants situated in the Rheb switch 2 segment, Y67A/I69A and I76A/D77A, exhibit a near total loss of function, whereas extensive replacement of the switch 1 segment and other surface residues with alanines causes relatively little disturbance of Rheb rescue of S6K1 from amino acid withdrawal. This is surprising in view of the minimal impact of guanyl nucleotide on Rheb switch 2 configuration. The loss of function Rheb switch 2 mutants are well expressed and exhibit partial agonist function in amino acid-replete cells. They are unimpaired in their ability to bind GTP or mammalian (m)TOR in vivo or in vitro, and the mTOR polypeptides retrieved with these inactive Rheb mutants exhibit kinase activity in vitro comparable with mTOR bound to wild type Rheb. We conclude that Rheb signaling to mTOR in vivo requires a Rheb switch 2-dependent interaction with an element other than the three known polypeptide components of TOR complex 1.


Subject(s)
Monomeric GTP-Binding Proteins/physiology , Neuropeptides/physiology , Protein Kinases/metabolism , Amino Acid Sequence , Animals , Cell Line , Guanosine Triphosphate/metabolism , Humans , Molecular Sequence Data , Monomeric GTP-Binding Proteins/chemistry , Monomeric GTP-Binding Proteins/metabolism , Mutation , Neuropeptides/chemistry , Peptides/chemistry , Ras Homolog Enriched in Brain Protein , Schizosaccharomyces/metabolism , Sequence Homology, Amino Acid , Signal Transduction , TOR Serine-Threonine Kinases , Transfection
13.
Am J Reprod Immunol ; 56(5-6): 275-85, 2006.
Article in English | MEDLINE | ID: mdl-17076671

ABSTRACT

OBJECTIVES: Soluble (s)HLA-G1 is produced by trophoblast cells. Aim was to analyze the capacities and mechanisms of sHLA-G1 to regulate interleukin (IL)-2-induced cytotoxicity of natural killer (NK) cells from human deciduas. METHODS: Natural killer cells were isolated from decidual layers of term placentae, stimulated or not with IL-2 and supplemented with various concentrations of recombinant soluble HLA-G1 (sHLA-G1). For NK cell cytotoxicity assays, K562 cells were used as targets. Expression of signal transducer and activator of transcription 3 (STAT3) and perforin was analyzed by Western blotting. Apoptosis was examined by assessment of poly(ADP-ribose) polymerase cleavage. NK cells were analyzed by flow cytometry for IL-2receptor-alpha (IL-2R alpha; CD25) and transferrin receptor CD71 expression. RESULTS: Interleukin-2 increases CD71, STAT3, perforin expression and cytotoxic potential of NK cells. Expression of CD71, STAT3 and perforin decreased simultaneously with cytotoxicity and dose-dependently when sHLA-G1 (1.6 micro g/mL-1.6 ng/mL) was added to IL-2 stimulated cultures. sHLA-G1 did not induce apoptosis and CD25 expression was not affected. CONCLUSION: Interleukin-2R alpha expression is not controlled by sHLA-G1, but its signal transducer STAT3 as well as several downstream effects, such as perforin expression, proliferation and cytotoxicity. The control of STAT3 bioavailability through sHLA-G1 may be a key regulator of the mentioned effects.


Subject(s)
Cytotoxicity, Immunologic/drug effects , Decidua/immunology , HLA Antigens/immunology , HLA Antigens/pharmacology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Term Birth/immunology , Adult , Antigens, CD/metabolism , Biomarkers , Cell Proliferation , Cell Separation , Cells, Cultured , Cytotoxicity, Immunologic/immunology , Decidua/drug effects , Female , HLA-G Antigens , Humans , Interleukin-2/pharmacology , Membrane Glycoproteins/metabolism , Perforin , Poly(ADP-ribose) Polymerases/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Receptors, Transferrin/metabolism , STAT3 Transcription Factor/metabolism , Solubility
14.
J Biol Chem ; 279(17): 17951-6, 2004 Apr 23.
Article in English | MEDLINE | ID: mdl-14963039

ABSTRACT

Antimicrobial peptides, such as LL-37, are found both in nonvertebrates and vertebrates, where they represent important components of innate immunity. Bacterial infections at epithelial surfaces are associated with substantial induction of LL-37 expression, which allows efficient lysis of the invading microbes. Peptide-mediated lysis results in the release of bacterial nucleic acids with potential pathobiological activity in the host. Here, we demonstrate that LL-37 targets extracellular DNA plasmid to the nuclear compartment of mammalian cells, where it is expressed. DNA transfer occurred at physiological LL-37 concentrations that killed bacterial cells, whereas virtually no cytotoxic or growth-inhibitory effects were observed in mammalian cells. Furthermore, LL-37 protected DNA from serum nuclease degradation. LL-37.DNA complex uptake was a saturable time- and temperature-dependent process and was sensitive to cholesterol-depleting agents that are known to disrupt lipid rafts and caveolae, as shown by flow cytometry. Confocal fluorescence microscopy studies showed localization of internalized DNA to compartments stained by cholera toxin B, a marker of lipid rafts, but failed to demonstrate any co-localization of internalized DNA with caveolin-positive endocytotic vesicles. Moreover, LL-37-mediated plasmid uptake and reporter gene expression were strictly dependent on cell surface proteoglycans. We conclude that the human antimicrobial peptide LL-37 binds to, protects, and efficiently targets DNA plasmid to the nuclei of mammalian cells through caveolae-independent membrane raft endocytosis and cell surface proteoglycans.


Subject(s)
Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Endocytosis , Gene Transfer Techniques , Proteoglycans/metabolism , Animals , CHO Cells , COS Cells , Cathelicidins , Cell Division , Cell Line , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cricetinae , DNA/chemistry , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Luciferases/metabolism , Membrane Microdomains/chemistry , Microscopy, Confocal , Microscopy, Fluorescence , Models, Biological , Peptides/chemistry , Plasmids/metabolism , Proteoglycans/chemistry , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL