Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Exp Mol Pathol ; 97(1): 154-65, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24984270

ABSTRACT

Mucoepidermoid carcinoma (MEC) is the most prevalent malignant tumor in major and minor salivary glands (SGs). We have recently identified human cytomegalovirus (hCMV) as a principle component in the multifactorial causation of SG-MEC. This finding is corroborated by the ability of the purified mouse CMV (mCMV) to induce malignant transformation of SG cells in a three-dimensional in vitro mouse model, using a similar oncogenic signaling pathway. Our prior studies indicate that the core tumor microenvironment (TME) is a key regulator of pathologic progression, particularly the cancer-associated fibroblast (CAF) component. Studies of early CAFs immunodetect aberrant expression of ECM components, as well as multiple growth factors, cytokines and transcription factors. Here we present the mechanistic insight derived from a mathematical structure ("wiring diagram") used to model complex relationships between a highly relevant (p=9.43×10(-12)) global "cancer network" of 32 genes and their known links. Detailed characterization of the functional architecture of the examined "cancer network" exposes the critical crosstalk and compensatory pathways that limit the efficacy of targeted anti-kinase therapies.


Subject(s)
Cytomegalovirus/pathogenicity , Gene Expression Regulation , Models, Theoretical , Salivary Glands/pathology , Salivary Glands/virology , Animals , Carcinoma, Mucoepidermoid/genetics , Carcinoma, Mucoepidermoid/pathology , Carcinoma, Mucoepidermoid/virology , Cytomegalovirus Infections/genetics , Epistasis, Genetic , Female , Gefitinib , Gene Regulatory Networks , Humans , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Salivary Gland Neoplasms/genetics , Salivary Gland Neoplasms/pathology , Salivary Gland Neoplasms/virology , Salivary Glands/physiopathology , Signal Transduction/drug effects , Tumor Microenvironment
2.
Exp Mol Pathol ; 94(2): 386-97, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23399805

ABSTRACT

Mucoepidermoid carcinoma (MEC) is the most common malignant tumor originating in major and minor salivary glands (SGs). Although the precise multifactorial etiology of human SG-MEC is largely unknown, we have recently shown that cytomegalovirus (CMV) is an important component of MEC tumorigenesis. Despite the well-documented overexpression of the EGFR → ERK signaling pathway in SG-MEC, there has been limited to no clinical success with inhibition of this pathway. Using our previously characterized mouse model of CMV-induced SG dysplasia/neoplasia, we report that inhibitors of the EGFR → ERK pathway do not ameliorate or rescue well-established pathology, either singly or in combination, but they do inhibit the evolution of progressive pathogenesis ("disease tolerance") in the face of mounting CMV burden. Failure to rescue SG pathology, suggested a possible increase in the ligand levels of alternative pathways that share cell proliferation and survival effectors (e.g. ERK and PI3K). Here we present evidence of a highly significant upregulation of ligands for the EGFR, FGFR, IL-6R, and TNFR signaling pathways, all of which converge upon the Raf/MEK/ERK amplifier module. This explains our finding that even in the presence of the highest nontoxic dose of an ERK phosphorylation inhibitor, pERK is undiminished. Given the considerable pathway crosstalk, a deep understanding of subversion and dysregulation of the SG interactome by CMV is a priori quite daunting. Circumventing this dilemma, we present evidence that concurrent inhibition of ERK phosphorylation (U0126) and CMV replication (acyclovir) obviates progressive pathogenesis and results in complete SG rescue (tumor regression). These findings provide a mechanistic foundation for potential clinical trials that utilize similar concurrent treatment with extant FDA-approved drugs.


Subject(s)
Carcinoma, Mucoepidermoid/virology , Extracellular Signal-Regulated MAP Kinases/metabolism , Herpesviridae Infections/pathology , Muromegalovirus/physiology , Submandibular Gland Neoplasms/virology , Submandibular Gland/pathology , Submandibular Gland/virology , Acyclovir/pharmacology , Amphiregulin , Animals , Carcinoma, Mucoepidermoid/metabolism , Carcinoma, Mucoepidermoid/pathology , Cytopathogenic Effect, Viral , Disease Models, Animal , EGF Family of Proteins , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Female , Fibroblast Growth Factor 8/metabolism , Glycoproteins/metabolism , Herpesviridae Infections/metabolism , Herpesviridae Infections/virology , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-6/metabolism , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Muromegalovirus/drug effects , Organ Culture Techniques , Phosphorylation , Submandibular Gland/metabolism , Submandibular Gland Neoplasms/metabolism , Submandibular Gland Neoplasms/pathology , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
3.
Herpesviridae ; 4(1): 1, 2013 Jan 23.
Article in English | MEDLINE | ID: mdl-23342981

ABSTRACT

BACKGROUND: Recently we identified a relationship between human cytomegalovirus (hCMV) and human salivary gland (SG) mucoepidermoid carcinoma (MEC) in over 90% of cases; tumorigenesis in these cases uniformly correlated with active hCMV protein expression and an upregulation of the EGFR → ERK pathway. Our previously characterized, novel mouse organ culture model of mouse CMV (mCMV)-induced tumorigenesis displays a number of histologic and molecular characteristics similar to human MEC. METHODS: Newborn mouse submandibular glands (SMGs) were incubated with 1 × 105 PFU/ml of lacZ-tagged mCMV RM427+ on day 0 for 24 hours and then cultured in virus-free media for a total of 6 or 12 days with or without EGFR/ERK inhibitors and/or aciclovir. SMGs were collected for histology, immunolocalization (pERK, FN, IL-6), viral distribution, or Western blot analysis (pERK). RESULTS: Here we report: (1) mouse SMG tumors soon exhibit an acquired resistance to EGFR/ERK pathway kinase inhibitors, alone or in combination; (2) long term tumor regression can only be sustained by concurrent inhibitor and antiviral treatment; (3) CMV-dependent, kinase inhibitor resistance is associated with overexpression of fibronectin and IL-6 proteins in abnormal stromal cells. CONCLUSIONS: Acquired resistance to kinase inhibitors is dependent upon CMV dysregulation of alternative pathways with downstream effectors common with the targeted pathway, a phenomenon with important therapeutic implications for human MEC of salivary glands.

4.
Proc Natl Acad Sci U S A ; 109(37): E2457-65, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22895791

ABSTRACT

The circadian clock controls many physiological parameters including immune response to infectious agents, which is mediated by activation of the transcription factor NF-κB. It is widely accepted that circadian regulation is based on periodic changes in gene expression that are triggered by transcriptional activity of the CLOCK/BMAL1 complex. Through the use of a mouse model system we show that daily variations in the intensity of the NF-κB response to a variety of immunomodulators are mediated by core circadian protein CLOCK, which can up-regulate NF-κB-mediated transcription in the absence of BMAL1; moreover, BMAL1 counteracts the CLOCK-dependent increase in the activation of NF-κB-responsive genes. Consistent with its regulatory function, CLOCK is found in protein complexes with the p65 subunit of NF-κB, and its overexpression correlates with an increase in specific phosphorylated and acetylated transcriptionally active forms of p65. In addition, activation of NF-κB in response to immunostimuli in mouse embryonic fibroblasts and primary hepatocytes isolated from Clock-deficient mice is significantly reduced compared with WT cells, whereas Clock-Δ19 mutation, which reduces the transactivation capacity of CLOCK on E-box-containing circadian promoters, has no effect on the ability of CLOCK to up-regulate NF-κB-responsive promoters. These findings establish a molecular link between two essential determinants of the circadian and immune mechanisms, the transcription factors CLOCK and NF-κB, respectively.


Subject(s)
CLOCK Proteins/metabolism , Circadian Rhythm/physiology , Gene Expression Regulation/immunology , Gene Expression Regulation/physiology , Transcription Factor RelA/metabolism , Transcription, Genetic/physiology , Analysis of Variance , Animals , Blotting, Western , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/genetics , Humans , Immunoprecipitation , Luciferases , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Peptides , Transcription, Genetic/genetics
5.
Virology ; 425(1): 53-60, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22284893

ABSTRACT

The NF-kB family of transcription factors regulates important biological functions including cell growth, survival and the immune response. We found that Human Papillomavirus type 16 (HPV-16) E7 and E6/E7 proteins inhibited basal and TNF-alpha-inducible NF-kB activity in human epithelial cells cultured from the cervical transformation zone, the anatomic region where most cervical cancers develop. In contrast, HPV-16 E6 regulated NF-kB in a cell type- and cell growth-dependent manner. NF-kB influenced immortalization of cervical cells by HPV16. Inhibition of NF-kB by an IkB alpha repressor mutant increased colony formation and immortalization by HPV-16. In contrast, activation of NF-kB by constitutive expression of p65 inhibited proliferation and immortalization. Our results suggest that inhibition of NF-kB by HPV-16 E6/E7 contributes to immortalization of cells from the cervical transformation zone.


Subject(s)
Cell Transformation, Viral , Human papillomavirus 16/pathogenicity , NF-kappa B/antagonists & inhibitors , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Repressor Proteins/metabolism , Cells, Cultured , Cervix Uteri/cytology , Cervix Uteri/metabolism , Cervix Uteri/virology , Epithelial Cells/metabolism , Epithelial Cells/virology , Female , Foreskin/cytology , Foreskin/virology , Gene Expression Regulation , Human papillomavirus 16/genetics , Human papillomavirus 16/growth & development , Human papillomavirus 16/metabolism , Humans , Male , NF-kappa B/metabolism , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Repressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...