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










Publication year range
1.
PLoS One ; 15(7): e0236741, 2020.
Article in English | MEDLINE | ID: mdl-32730300

ABSTRACT

Aryl hydrocarbon receptor (AHR) agonists such as dioxin have been associated with obesity and the development of diabetes. Whole-body Ahr knockout mice on high-fat diet (HFD) have been shown to resist obesity and hepatic steatosis. Tissue-specific knockout of Ahr in mature adipocytes via adiponectin-Cre exacerbates obesity while knockout in liver increases steatosis without having significant effects on obesity. Our previous studies demonstrated that treatment of subcutaneous preadipocytes with exogenous or endogenous AHR agonists disrupts maturation into functional adipocytes in vitro. Here, we used platelet-derived growth factor receptor alpha (Pdgfrα)-Cre mice, a Cre model previously established to knock out genes in preadipocyte lineages and other cell types, but not liver cells, to further define AHR's role in obesity. We demonstrate that Pdgfrα-Cre Ahr-floxed (Ahrfl/fl) knockout mice are protected from HFD-induced obesity compared to non-knockout Ahrfl/fl mice (control mice). The Pdgfrα-Cre Ahrfl/fl knockout mice were also protected from increased adiposity, enlargement of adipocyte size, and liver steatosis while on the HFD compared to control mice. On a regular control diet, knockout and non-knockout mice showed no differences in weight gain, indicating the protective phenotype arises only when animals are challenged by a HFD. At the cellular level, cultured cells from brown adipose tissue (BAT) of Pdgfrα-Cre Ahrfl/fl mice were more responsive than cells from controls to transcriptional activation of the thermogenic uncoupling protein 1 (Ucp1) gene by norepinephrine, suggesting an ability to burn more energy under certain conditions. Collectively, our results show that knockout of Ahr mediated by Pdgfrα-Cre is protective against diet-induced obesity and suggest a mechanism by which enhanced UCP1 activity within BAT might confer these effects.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Diet, High-Fat/adverse effects , Fatty Liver/prevention & control , Integrases/metabolism , Obesity/prevention & control , Receptor, Platelet-Derived Growth Factor alpha/physiology , Receptors, Aryl Hydrocarbon/physiology , Adiposity , Animals , Energy Metabolism , Fatty Liver/etiology , Fatty Liver/pathology , Female , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/etiology , Obesity/pathology , Thermogenesis
2.
Cells ; 9(3)2020 02 25.
Article in English | MEDLINE | ID: mdl-32106632

ABSTRACT

BACKGROUND: Development of radioresistance in oral squamous cell carcinoma (OSCC) remains a significant problem in cancer treatment, contributing to the lack of improvement in survival trends in recent decades. Effective strategies to overcome radioresistance are necessary to improve the therapeutic outcomes of radiotherapy in OSCC patients. METHODS: Cells and xenograft tumors were irradiated using the Small Animal Radiation Research Platform. AKT inhibitor capivasertib (AZD5363) was encapsulated into cathepsin B-responsible nanoparticles (NPs) for tumor-specific delivery. Cell viability was measured by alamarBlue, cell growth was determined by colony formation and 3D culture, and apoptosis was assessed by flow cytometry with the staining of Fluorescein isothiocyanate (FITC) Annexin V and PI. An orthotopic tongue tumor model was used to evaluate the in vivo therapeutic effects. The molecular changes induced by the treatments were assessed by Western blotting and immunohistochemistry. RESULTS: We show that upregulation of AKT signaling is the critical mechanism for radioresistance in OSCC cells, and AKT inactivation by a selective and potent AKT inhibitor capivasertib results in radiosensitivity. Moreover, relative to irradiation (IR) alone, IR combined with the delivery of capivasertib in association with tumor-seeking NPs greatly enhanced tumor cell repression in 3D cell cultures and OSCC tumor shrinkage in an orthotopic mouse model. CONCLUSIONS: These data indicate that capivasertib is a potent agent that sensitizes radioresistant OSCC cells to IR and is a promising strategy to overcome failure of radiotherapy in OSCC patients.


Subject(s)
Mouth Neoplasms/diet therapy , Nanoparticles/metabolism , Proto-Oncogene Proteins c-akt/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Animals , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred NOD , Mouth Neoplasms/radiotherapy , Proto-Oncogene Proteins c-akt/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Xenograft Model Antitumor Assays
3.
Head Neck Pathol ; 13(4): 635-642, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30888638

ABSTRACT

Proliferative verrucous leukoplakia (PVL) is a premalignant condition of the oral mucosa with > 70% chance of progression to squamous cell carcinoma (SCC), while lacking the common risks and behavior seen in non-PVL oral squamous carcinogenesis. PVL follows a multi-stage slow, relentless and usually multifocal expansion of surface epithelial thickening that over time takes on a verrucous architecture, eventually leading to verrucous carcinoma and/or dysplasia followed by "conventional" SCC, a process that takes years and is notoriously difficult to manage. As mucosal surfaces and carcinomas arising at these sites, are colonized by microorganisms, host receptors for microbial products have received attention as potential contributors to carcinogenesis. Studies show that microbial pattern recognition toll-like receptor (TLR)2 in various epithelial cells is upregulated in premalignant lesions and in malignant cells and can activate oncogenic pathways. Because of the highly progressive nature of PVL, we examined TLR2 expression in well-characterized PVL samples by immunohistochemistry. We found that, similar to epithelial dysplasia and SCC, PVL keratinocytes throughout the epithelial thickness showed diffuse TLR2 expression even in early stage lesions prior to onset of dysplasia. In contrast, oral mucosal samples in the absence of hyperorothokeratosis or dysplasia, expressed TLR2 primarily in the basal and parabasal layers. Given the high rates of PVL transformation and the previously established pro-cancer role of high TLR2 expression in malignant oral squamous cells, it is important to determine how its' expression and functions are regulated in the oral squamous epithelium, and what is the specific TLR2 role in carcinogenesis.


Subject(s)
Biomarkers, Tumor/analysis , Leukoplakia, Oral/pathology , Squamous Cell Carcinoma of Head and Neck/pathology , Toll-Like Receptor 2/biosynthesis , Aged , Aged, 80 and over , Female , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Leukoplakia, Oral/metabolism , Male , Middle Aged , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Squamous Cell Carcinoma of Head and Neck/metabolism
4.
Oncotarget ; 9(6): 6814-6829, 2018 Jan 23.
Article in English | MEDLINE | ID: mdl-29467931

ABSTRACT

Patient treatment for oral squamous cell carcinoma (OSCC) not associated with Human papillomavirus remains problematic. OSCC microenvironment is typically inflamed and colonized by microorganisms, providing ligands for toll-like receptors (TLR). In immune cells TLR2 and TLR4 activate NF-kB and extracellular signal regulated kinase (ERK)1/2 pathways, leading to upregulation of inhibitory adenosine receptors A2a and A2b, and reduction in stimulatory A1 and A3. How TLR and adenosine receptors function in SCC cells is not understood. To address this gap, we evaluated TLR and adenosine receptor expression and function in human OSCC cells and keratinocytes. TLR2 and A2a were co-expressed in pre-cancer and SCC cells of 17 oral specimens. In vitro, 5/6 OSCC lines expressed more TLR2 than TLR1, 4 or 6 mRNA. TLR2 ligands stimulated A2a expression in TLR2-high cell lines, but no A1 or A3 was detected with or without stimuli. In TLR2-high OSCC, TLR2/1, 2/6 and adenosine receptor agonists activated ERK1/2. TLR2-mediated ERK1/2 phosphorylation resulted in accumulation of c-FOS, ERK-dependent cell proliferation and reduced caspase-3 activity. Similar ERK1/2-dependent proliferation and decreased caspase-3 activity were caused by combined TLR2 and adenosine receptor stimuli. We conclude that TLR2 and adenosine receptor agonists, known to be present in the tumor microenvironment, may contribute to OSCC progression in part via direct effects on the ERK1/2 pathway in squamous carcinoma cells.

5.
J Immunol ; 200(5): 1781-1789, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29386257

ABSTRACT

At mucosal sites such as the intestine, the immune system launches robust immunity against invading pathogens while maintaining a state of tolerance to commensal flora and ingested food Ags. The molecular mechanisms underlying this phenomenon remain poorly understood. In this study, we report that signaling by GPR81, a receptor for lactate, in colonic dendritic cells and macrophages plays an important role in suppressing colonic inflammation and restoring colonic homeostasis. Genetic deletion of GPR81 in mice led to increased Th1/Th17 cell differentiation and reduced regulatory T cell differentiation, resulting in enhanced susceptibility to colonic inflammation. This was due to increased production of proinflammatory cytokines (IL-6, IL-1ß, and TNF-α) and decreased expression of immune regulatory factors (IL-10, retinoic acid, and IDO) by intestinal APCs lacking GPR81. Consistent with these findings, pharmacological activation of GPR81 decreased inflammatory cytokine expression and ameliorated colonic inflammation. Taken together, these findings identify a new and important role for the GPR81 signaling pathway in regulating immune tolerance and colonic inflammation. Thus, manipulation of the GPR81 pathway could provide novel opportunities for enhancing regulatory responses and treating colonic inflammation.


Subject(s)
Colitis/metabolism , Homeostasis/physiology , Lactic Acid/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cytokines/metabolism , Disease Models, Animal , Female , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Th1 Cells/metabolism
6.
Article in English | MEDLINE | ID: mdl-27260276

ABSTRACT

Oral lichen planus is a noninfectious, chronic inflammatory condition that involves the oral mucosal stratified squamous epithelium and the underlying lamina propria and may be accompanied by skin lesions. This overview describes the current understanding of the immunopathologic mechanisms implicated in oral lichen planus.


Subject(s)
Lichen Planus, Oral/etiology , Lichen Planus, Oral/immunology , Lichen Planus, Oral/pathology , Humans , Risk Factors
7.
PLoS Pathog ; 10(2): e1004647, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25679217

ABSTRACT

Signaling via pattern recognition receptors (PRRs) expressed on professional antigen presenting cells, such as dendritic cells (DCs), is crucial to the fate of engulfed microbes. Among the many PRRs expressed by DCs are Toll-like receptors (TLRs) and C-type lectins such as DC-SIGN. DC-SIGN is targeted by several major human pathogens for immune-evasion, although its role in intracellular routing of pathogens to autophagosomes is poorly understood. Here we examined the role of DC-SIGN and TLRs in evasion of autophagy and survival of Porphyromonas gingivalis in human monocyte-derived DCs (MoDCs). We employed a panel of P. gingivalis isogenic fimbriae deficient strains with defined defects in Mfa-1 fimbriae, a DC-SIGN ligand, and FimA fimbriae, a TLR2 agonist. Our results show that DC-SIGN dependent uptake of Mfa1+P. gingivalis strains by MoDCs resulted in lower intracellular killing and higher intracellular content of P. gingivalis. Moreover, Mfa1+P. gingivalis was mostly contained within single membrane vesicles, where it survived intracellularly. Survival was decreased by activation of TLR2 and/or autophagy. Mfa1+P. gingivalis strain did not induce significant levels of Rab5, LC3-II, and LAMP1. In contrast, P. gingivalis uptake through a DC-SIGN independent manner was associated with early endosomal routing through Rab5, increased LC3-II and LAMP-1, as well as the formation of double membrane intracellular phagophores, a characteristic feature of autophagy. These results suggest that selective engagement of DC-SIGN by Mfa-1+P. gingivalis promotes evasion of antibacterial autophagy and lysosome fusion, resulting in intracellular persistence in myeloid DCs; however TLR2 activation can overcome autophagy evasion and pathogen persistence in DCs.


Subject(s)
Autophagy/immunology , Cell Adhesion Molecules/metabolism , Dendritic Cells/metabolism , Lectins, C-Type/metabolism , Myeloid Cells/metabolism , Porphyromonas gingivalis/metabolism , Receptors, Cell Surface/metabolism , Toll-Like Receptor 2/metabolism , Dendrites/ultrastructure , Dendritic Cells/immunology , Dendritic Cells/ultrastructure , Fimbriae, Bacterial , Humans , Intracellular Space/immunology , Intracellular Space/metabolism , Monocytes/immunology , Monocytes/ultrastructure , Myeloid Cells/immunology , Toll-Like Receptor 2/immunology
8.
N Y State Dent J ; 79(3): 46-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23767401

ABSTRACT

Many lesions in the oral cavity may present with a papillary or pebbly clinical appearance. Although the great majority of these papillary lesions are histologically diagnosed as squamous or viral papillomas, there are occasional cases of other more unusual possibilities. Angiokeratomas are uncommon vascular lesions that often present clinically as papillomas. They may also present with other varying clinical appearances that range from pigmented lesions to hemangiomas. However, all forms demonstrate a characteristic microscopic appearance consisting of hyperkeratotic, hyperplastic epithelium covering connective tissue with abundant blood vessels that are sharply confined to the connective tissue papillae. Angiokeratomas generally involve the skin and are often associated with an underlying systemic metabolic disease such as Fabry's disease or fucosidosis. Patients with these systemic diseases may have multiple lesions, with possible involvement of the oral mucosa. However, solitary lesions involving only the oral cavity are rare; only eight previous cases have been documented. In this paper, we describe a case of solitary angiokeratoma presenting as a papillary lesion on the ventral tongue of an 18-year-old male. The lesion was surgically excised and no recurrence has been reported to date. Although this patient had no other lesions or systemic issues, we stress the importance of evaluating a patient with a diagnosis of angiokeratoma of the oral cavity for underlying systemic metabolic disease.


Subject(s)
Angiokeratoma/pathology , Tongue Neoplasms/pathology , Adolescent , Angiokeratoma/surgery , Diagnosis, Differential , Humans , Laser Therapy , Male , Papilloma/diagnosis , Tongue Neoplasms/surgery
9.
Cancer Microenviron ; 6(1): 41-55, 2013 Apr.
Article in English | MEDLINE | ID: mdl-21809059

ABSTRACT

In the inflammatory mucosal microenvironment of head and neck SCC (HNSCC), DC express CD16 and are usually in direct contact with tumor cells. Mucosal and inflammation-associated DC develop from monocytes, and monocyte-derived DC are used in HNSCC immunotherapy. However, beyond apoptotic tumor cell uptake and presentation of tumor antigens by DC, HNSCC cell interactions with DC are poorly understood. Using co-cultures of monocyte-derived DC and two established HNSCC cell lines that represent well- and poorly-differentiated SCC, respectively, we found that carcinoma cells induced significant increases in CD16 expression on DC while promoting a CD1a(+)CD86(dim) immature phenotype, similar to that observed in HNSCC specimens. Moreover, HNSCC cells affected steady-state and CCL21-induced migration of DC, and these effects were donor-dependent. The CCL21-induced migration directly correlated with HNSCC-mediated effects on CCR7 and CD38 expression on DC-SIGN-high DC. The dominant pattern seen in six out of nine donors was the increase in steady-state and CCL21-induced DC migration in co-cultures with HNSCC, while the reverse pattern, i.e., decreased DC migration in co-cultures with SCC, was identified in two donors. A split in migratory DC behavior, i.e. increase with one HNSCC cell line and a decrease with the second cell line, was observed in one donor. Remarkably, the numbers of live detached HNSCC cells were orders of magnitude higher in DC-HNSCC co-cultures than in parallel HNSCC cell cultures without DC. This study provides novel insights into the effects of DC-HNSCC interactions relevant to the tumor microenvironment.

10.
Int J Antimicrob Agents ; 40(1): 75-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22578747

ABSTRACT

Human ß-defensin-3 (HBD3) is a small, cationic, host defence peptide with broad antimicrobial activities and diverse innate immune functions. HBD3 binds to many microbial antigens and, in this study, we hypothesised that the known binding of HBD3 to Porphyromonas gingivalis recombinant haemagglutinin B (rHagB) alters, but does not inhibit, the binding of rHagB to human dendritic cells. To test this, human myeloid dendritic cells were incubated for 5 min with rHagB, HBD3 + rHagB (10:1 molar ratio), HBD3 or 0.1 M phosphate-buffered saline (PBS) (pH 7.2) and were then rapidly fixed and processed for confocal microscopy and ultramicrotomy. rHagB and HBD3 could be detected with primary monoclonal mouse antibody to rHagB (MoAb 1858) or polyclonal rabbit antibody to HBD3 (P241) and secondary fluorescent-labelled anti-mouse or anti-rabbit antibodies (confocal microscopy) or protein A-colloidal gold (immunoelectron microscopy). In cells incubated with rHagB only, fluorescence and protein A-colloidal gold were seen at the cell surface and throughout the cytoplasm. In cells incubated with HBD3+rHagB, fluorescence was observed only at the cell surface in a 'string of pearls' configuration. Overall, these results suggest that HBD3 binding to rHagB alters, but does not inhibit, the binding of rHagB to human myeloid dendritic cells.


Subject(s)
Adhesins, Bacterial/metabolism , Anti-Infective Agents/metabolism , Dendritic Cells/microbiology , Porphyromonas gingivalis/pathogenicity , beta-Defensins/metabolism , Cells, Cultured , Humans , Lectins/metabolism , Microscopy, Confocal , Microscopy, Immunoelectron , Porphyromonas gingivalis/metabolism , Protein Binding
11.
Inflammation ; 33(4): 207-23, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20084448

ABSTRACT

Bacteria and chronic inflammation are present in squamous cell carcinoma of the head and neck (HNSCC), but their roles in the pathogenesis of HNSCC are unclear. Our studies described here revealed that human monocytes co-cultured short term with HNSCC cells were more likely to express CD16, and CD16(+) small mononuclear cells were common in HNSCC specimens. In addition, we identified monocytes as the primary source of LPS-induced IL-6 and TNF-alpha in the monocyte-HNSCC co-cultures. Remarkably, relative to LPS-stimulated monocytes cultured alone, HNSCC cells profoundly suppressed LPS-induced TNF-alpha in monocytes, without compromising IL-6 production. High levels of cytoprotective factors like IL-6 and low levels of TNF-alpha are important for the tumor microenvironment that enables tumor cell survival, affects monocyte differentiation and may contribute to tumor colonization by bacteria. This study provides novel observations that HNSCC cells affect monocyte phenotype and function, which are relevant to the regulation of the HNSCC microenvironment.


Subject(s)
Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/pathology , Lipopolysaccharides/pharmacology , Monocytes/immunology , Receptors, IgG/immunology , Tumor Necrosis Factor-alpha/metabolism , Carcinoma, Squamous Cell/immunology , Cell Line, Tumor/cytology , Cells, Cultured/cytology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Coculture Techniques , Disease Progression , GPI-Linked Proteins , Head and Neck Neoplasms/immunology , Humans , Inflammation , Interleukin-6/biosynthesis , Interleukin-6/metabolism , Keratinocytes/cytology , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Mouth Mucosa/pathology , Phagocytosis , Phenotype , Receptors, IgG/biosynthesis , Stomatitis/pathology
12.
Immunol Cell Biol ; 86(8): 643-9, 2008.
Article in English | MEDLINE | ID: mdl-18711400

ABSTRACT

Regulatory mechanisms in mucosal secretions and tissues recognize antigens and attenuate pro-inflammatory cytokine responses. Here, we asked whether human beta-defensin 3 (HBD3) serves as an upstream suppressor of cytokine signaling that binds and attenuates pro-inflammatory cytokine responses to recombinant hemagglutinin B (rHagB), a non-fimbrial adhesin from Porphyromonas gingivalis strain 381. We found that HBD3 binds to immobilized rHagB and produces a significantly higher resonance unit signal in surface plasmon resonance spectroscopic analysis, than HBD2 and HBD1 that are used as control defensins. Furthermore, we found that HBD3 significantly attenuates (P<0.05) the interleukin (IL)-6, IL-10, granulocyte macrophage colony stimulating factor (GM-CSF) and tumor-necrosis factor-alpha (TNF-alpha) responses induced by rHagB in human myeloid dendritic cell culture supernatants and the extracellular signal-regulated kinases (ERK 1/2) response in human myeloid dendritic cell lysates. Thus, HBD3 binds rHagB and this interaction may be an important initial step to attenuate a pro-inflammatory cytokine response and an ERK 1/2 response.


Subject(s)
Adhesins, Bacterial/metabolism , Cytokines/metabolism , Dendritic Cells/immunology , Immunity, Innate , Porphyromonas gingivalis/immunology , beta-Defensins/metabolism , Adhesins, Bacterial/immunology , Cytokines/immunology , Dendritic Cells/metabolism , Extracellular Signal-Regulated MAP Kinases , Humans , Lectins/immunology , Lectins/metabolism , MAP Kinase Signaling System , Porphyromonas gingivalis/drug effects , Recombinant Proteins/metabolism , Surface Plasmon Resonance , beta-Defensins/immunology
13.
Head Neck Pathol ; 2(1): 1-12, 2008 Mar.
Article in English | MEDLINE | ID: mdl-19603082

ABSTRACT

Oral and oro-pharyngeal squamous cell carcinomas (OSCC) exhibit surface breach, and recent studies have demonstrated bacterial contamination of primary and metastatic OSCC. Increasing concentrations of inflammatory products, such as interleukin (IL)-6 and vascular endothelial growth factor (VEGF), correlate with, and contribute to, cancer progression, but their regulation in OSCC is poorly understood. We hypothesized that monocyte-lineage cells and bacterial contamination may contribute important inflammatory products that can support OSCC progression. We found that relative to non-specific chronic mucositis, oral carcinoma-in-situ/superficially-invasive OSCC contained more monocyte-lineage cells. In vitro, we used lipopolysaccharide (LPS) to model bacterial contamination, and evaluated the effects of oral and oropharyngeal (O)SCC-monocyte interactions and of LPS on OSCC cells and on the production of IL-6 and VEGF. OSCC cell lines varied in constitutive cytokine and chemokine production, and OSCC-monocyte interactions in the absence of LPS stimulated IL-6 and VEGF occasionally, while LPS-OSCC-monocyte interactions were always strongly stimulatory. Importantly, LPS independently stimulated some OSCC lines to secrete monocyte-dendritic cell chemoattractants CCL2 and/or CCL20, as well as IL-6 and/or VEGF. While very little constitutive Y705-STAT3 phosphorylation (pY705-STAT3) was detectable in HNSCC lines, IL-6 rapidly induced pY705-STAT3 in OSCC lines that produced little IL-6 constitutively. Supernatants from LPS-OSCC-monocyte co-cultures always rapidly and strongly activated STAT3, which was partly due to IL-6. We conclude that monocytes and microbial contamination have the potential to contribute to OSCC progression, as STAT3 activation in OSCC cells depends on soluble factors, which are consistently available through LPS-OSCC-monocyte interactions.


Subject(s)
Carcinoma in Situ/metabolism , Carcinoma, Squamous Cell/metabolism , Lipopolysaccharides/pharmacology , Monocytes/metabolism , Mouth Neoplasms/metabolism , Oropharyngeal Neoplasms/metabolism , STAT3 Transcription Factor/biosynthesis , Carcinoma in Situ/drug therapy , Carcinoma in Situ/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Coculture Techniques , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dendritic Cells/pathology , Flow Cytometry , Humans , Interleukin-6/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Monocytes/drug effects , Monocytes/pathology , Mouth Neoplasms/drug therapy , Mouth Neoplasms/pathology , Oropharyngeal Neoplasms/drug therapy , Oropharyngeal Neoplasms/pathology , Vascular Endothelial Growth Factor A/metabolism
14.
Cancer Immunol Immunother ; 52(2): 107-15, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12594574

ABSTRACT

Tumor cells stimulate natural killer (NK) cell effector functions, but the regulation of cytokine secretion and cytolysis is incompletely understood. We tested whether oral and pharyngeal squamous cell carcinoma cell lines differentially stimulated NK cell interferon-gamma (IFN-gamma) secretion and cytolysis using a clone of the NK-92-transformed human NK cell line, NK92.35. SCC-4 and SCC-25 cells, but not FaDu or Cal 27 cells, stimulated robust NK92.35 IFN-gamma secretion. All four carcinoma cell lines were lysed by NK92.35 cells. These findings indicate that carcinoma cells differentially stimulate NK cell IFN-gamma secretion and cytolysis. In Transwell experiments, a combination of SCC-4 or SCC-25 cell soluble factors and contact with FaDu cells synergistically stimulated NK92.35 cell IFN-gamma secretion. Stimulatory SCC-4 cells constitutively secreted IL-18, a cytokine that potently augments IFN-gamma secretion by T cells and NK cells. In contrast, poorly stimulatory FaDu cells produced little or no IL-18, but synergized with recombinant IL-18 to stimulate NK92.35 IFN-gamma secretion. mAb to IL-18 or IL-18 receptor diminished SCC-4-stimulated IFN-gamma secretion by NK92.35 cells and by nontransformed NK cells. Thus, IL-18 was necessary for optimal carcinoma stimulation of NK cell IFN-gamma secretion. In vivo, oral and upper aerodigestive tract epithelia and carcinomas produced IL-18, but one squamous cell carcinoma had heterogeneous IL-18 expression. Thus IL-18 production can account for squamous cell carcinoma differential stimulation of NK cell effector functions in vitro and may be important for stimulation of NK cells in vivo.


Subject(s)
Carcinoma, Squamous Cell/immunology , Interleukin-18/physiology , Killer Cells, Natural/immunology , Cytotoxicity, Immunologic , Humans , Interferon-gamma/metabolism , Tumor Cells, Cultured
15.
J Exp Med ; 197(2): 245-55, 2003 Jan 20.
Article in English | MEDLINE | ID: mdl-12538663

ABSTRACT

Killer immunoglobulin-like receptors (KIR) bind self-major histocompatibility complex class I molecules, allowing natural killer (NK) cells to recognize aberrant cells that have down-regulated class I. NK cells express variable numbers and combinations of highly homologous clonally restricted KIR genes, but uniformly express KIR2DL4. We show that NK clones express both 2DL4 alleles and either one or both alleles of the clonally restricted KIR 3DL1 and 3DL2 genes. Despite allele-independent expression, 3DL1 alleles differed in the core promoter by only one or two nucleotides. Allele-specific 3DL1 gene expression correlated with promoter and 5' gene DNA hypomethylation in NK cells in vitro and in vivo. The DNA methylase inhibitor, 5-aza-2'-deoxycytidine, induced KIR DNA hypomethylation and heterogeneous expression of multiple KIR genes. Thus, NK cells use DNA methylation to maintain clonally restricted expression of highly homologous KIR genes and alleles.


Subject(s)
DNA Methylation , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Receptors, Immunologic/genetics , Alleles , Cell Line , Clone Cells , Gene Expression , Histocompatibility Antigens Class I/metabolism , Humans , Receptors, KIR , Receptors, KIR2DL4 , Receptors, KIR3DL1 , Receptors, KIR3DL2
SELECTION OF CITATIONS
SEARCH DETAIL
...