Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Pain ; 7: 70, 2011 Sep 21.
Article in English | MEDLINE | ID: mdl-21936900

ABSTRACT

Neuropathic pain is a debilitating clinical condition with few efficacious treatments, warranting development of novel therapeutics. We hypothesized that dysregulated translation regulation pathways may underlie neuropathic pain. Peripheral nerve injury induced reorganization of translation machinery in the peripheral nervous system of rats and mice, including enhanced mTOR and ERK activity, increased phosphorylation of mTOR and ERK downstream targets, augmented eIF4F complex formation and enhanced nascent protein synthesis. The AMP activated protein kinase (AMPK) activators, metformin and A769662, inhibited translation regulation signaling pathways, eIF4F complex formation, nascent protein synthesis in injured nerves and sodium channel-dependent excitability of sensory neurons resulting in a resolution of neuropathic allodynia. Therefore, injury-induced dysregulation of translation control underlies pathology leading to neuropathic pain and reveals AMPK as a novel therapeutic target for the potential treatment of neuropathic pain.


Subject(s)
Molecular Targeted Therapy , Neuralgia/drug therapy , Neuralgia/enzymology , Protein Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Action Potentials/drug effects , Animals , Blotting, Western , Disease Models, Animal , Enzyme Activators/pharmacology , Enzyme Activators/therapeutic use , Eukaryotic Initiation Factor-4F/biosynthesis , Hyperalgesia/complications , Hyperalgesia/drug therapy , Hyperalgesia/pathology , Male , Mice , Mice, Inbred ICR , Neuralgia/physiopathology , Protein Biosynthesis/drug effects , Rats , Rats, Sprague-Dawley , Sciatic Nerve/drug effects , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/pathology , Signal Transduction/drug effects
2.
J Neurosci ; 30(45): 15113-23, 2010 Nov 10.
Article in English | MEDLINE | ID: mdl-21068317

ABSTRACT

Despite the emergence of translational control pathways as mediators of nociceptive sensitization, effector molecules and mechanisms responsible for modulating activity in these pathways in pain conditions are largely unknown. We demonstrate that two major algogens, the cytokine interleukin 6 (IL-6) and the neurotrophin nerve growth factor (NGF), which are intimately linked to nociceptive plasticity across preclinical models and human pain conditions, signal primarily through two distinct pathways to enhance translation in sensory neurons by converging onto the eukaryotic initiation factor (eIF) eIF4F complex. We directly demonstrate that the net result of IL-6 and NGF signaling is an enhancement of eIF4F complex formation and an induction of nascent protein synthesis in primary afferent neurons and their axons. Moreover, IL-6- and NGF-induced mechanical nociceptive plasticity is blocked by inhibitors of general and cap-dependent protein synthesis. These results establish IL-6- and NGF-mediated cap-dependent translation of local proteins as a new model for nociceptive plasticity.


Subject(s)
Eukaryotic Initiation Factor-4F/metabolism , Interleukin-6/metabolism , Nerve Growth Factor/metabolism , Neuronal Plasticity/physiology , Nociceptors/metabolism , Protein Biosynthesis/physiology , Signal Transduction/physiology , Analysis of Variance , Animals , Blotting, Western , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Immunohistochemistry , Interleukin-6/pharmacology , Male , Mice , Mice, Inbred ICR , Nerve Growth Factor/pharmacology , Neuronal Plasticity/drug effects , Nociceptors/drug effects , Phosphorylation , Protein Biosynthesis/drug effects , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
3.
PPAR Res ; 2008: 790568, 2008.
Article in English | MEDLINE | ID: mdl-18769553

ABSTRACT

Decreased expression of peroxisome proliferator activated receptor-gamma (PPARgamma) and high levels of the proinflammatory enzyme cyclooxygenase-2 (COX-2) have been observed in many tumor types. Both reduced (PPARgamma) expression and elevated COX-2 within the tumor are associated with poor prognosis in lung cancer patients, and recent work has indicated that these signaling pathways may be interrelated. Synthetic (PPARgamma) agonists such as the thiazolidinedione (TZD) class of anti-diabetic drugs can decrease COX-2 levels, inhibit growth of non-small-cell lung cancer (NSCLC) cells in vitro, and block tumor progression in xenograft models. TZDs alter the expression of COX-2 and consequent production of the protumorigenic inflammatory molecule prostaglandin E2 (PGE2) through both (PPARgamma) dependent and independent mechanisms. Certain TZDs also reduce expression of PGE2 receptors or upregulate the PGE2 catabolic enzyme 15-prostaglandin dehydrogenase. As several COX-2 enzymatic products have antitumor properties and specific COX-2 inhibition has been associated with increased risk of adverse cardiac events, directly reducing the effects or concentration of PGE2 may provide a more safe and effective strategy for lung cancer treatment. Understanding the mechanisms underlying these effects may be helpful for designing anticancer therapies. This article summarizes recent research on the relationship between (PPARgamma), TZDs, and the COX-2/PGE2 pathways in lung cancer.

4.
Crit Rev Oncol Hematol ; 66(3): 208-17, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18304833

ABSTRACT

Lung carcinogenesis is a complex process involving the acquisition of genetic mutations that confer cancer development and the malignant phenotype, and is critically linked to apoptosis resistance, unregulated proliferation, invasion, metastasis, and angiogenesis. Epithelial mesenchymal transition (EMT) in cancer is an unregulated process in a host environment with deregulated inflammatory response that impairs cell-mediated immunity and permits cancer progression. Given the immunosuppressive tumor environment, strategies to reverse these events by stimulating host immune responses are an important area of investigation. Cyclooxygenase 2 (COX-2) and its downstream signaling pathways are potential targets for lung cancer chemoprevention and therapy. Clinical trials are underway to evaluate COX-2 inhibitors as adjuvants to chemotherapy in patients with lung cancer and to determine efficacy in prevention of bronchogenic carcinoma. The understanding of molecular mechanisms involved in inflammation and lung carcinogenesis provide insight for new drug development that target reversible, non-mutational events in the chemoprevention and treatment of lung cancer.


Subject(s)
Cyclooxygenase 2 Inhibitors/therapeutic use , Cyclooxygenase 2/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , T-Lymphocyte Subsets/immunology , Chemotherapy, Adjuvant , Clinical Trials as Topic , Cytokines/immunology , Cytokines/metabolism , ErbB Receptors/metabolism , Genes, erbB-1 , Humans , Immune Tolerance , Inflammation , Lung/immunology , Lung/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/prevention & control , Signal Transduction , T-Lymphocyte Subsets/metabolism
5.
Expert Rev Anticancer Ther ; 7(10): 1405-21, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17944566

ABSTRACT

Lung carcinogenesis is a complex process requiring the acquisition of genetic mutations that confer the malignant phenotype as well as epigenetic alterations that may be manipulated in the course of therapy. Inflammatory signals in the lung cancer microenvironment can promote apoptosis resistance, proliferation, invasion, metastasis, and secretion of proangiogenic and immunosuppressive factors. Here, we discuss several prototypical inflammatory mediators controlling the malignant phenotype in lung cancer. Investigation into the detailed molecular mechanisms underlying the tumor-promoting effects of inflammation in lung cancer has revealed novel potential drug targets. Cytokines, growth factors and small-molecule inflammatory mediators released in the developing tumor microenvironment pave the way for epithelial-mesenchymal transition, the shift from a polarized, epithelial phenotype to a highly motile mesenchymal phenotype that becomes dysregulated during tumor invasion. Inflammatory mediators within the tumor microenvironment are derived from neoplastic cells as well as stromal and inflammatory cells; thus, lung cancer develops in a host environment in which the deregulated inflammatory response promotes tumor progression. Inflammation-related metabolic and catabolic enzymes (prostaglandin E(2) synthase, prostaglandin I(2) synthase and 15-hydroxyprostaglandin dehydrogenase), cell-surface receptors (E-type prostaglandin receptors) and transcription factors (ZEB1, SNAIL, PPARs, STATs and NF-kappaB) are differentially expressed in lung cancer cells compared with normal lung epithelial cells and, thus, may contribute to tumor initiation and progression. These newly discovered molecular mechanisms in the pathogenesis of lung cancer provide novel opportunities for targeted therapy and prevention in lung cancer.


Subject(s)
Cocarcinogenesis , Inflammation/complications , Lung Neoplasms/prevention & control , Animals , Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinogens, Environmental/adverse effects , Cell Differentiation/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Chronic Disease , Cyclooxygenase 2/physiology , Cyclooxygenase 2 Inhibitors/administration & dosage , Cyclooxygenase 2 Inhibitors/therapeutic use , Cytokines/physiology , Dinoprostone/metabolism , Epigenesis, Genetic , Humans , Inflammation/chemically induced , Inflammation/prevention & control , Lung Neoplasms/etiology , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Mice , Randomized Controlled Trials as Topic , Smoking/adverse effects , Tumor Escape
6.
Am J Respir Cell Mol Biol ; 37(5): 578-88, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17600311

ABSTRACT

Cyclooxygenase-2 (COX-2) is a key enzyme in the production of prostaglandins and thromboxanes from free arachidonic acid. Increasing evidence suggests that COX-2 plays a role in tumorigenesis. A variety of stimuli induce COX-2 and it is overexpressed in many tumors, including non-small cell lung cancer (NSCLC). We studied the regulation of COX-2 expression in immortalized human bronchial epithelial cells (HBECs) by transforming growth factor-beta1 (TGF-beta1) and epidermal growth factor (EGF) because these two growth factors are present in both the pulmonary milieu of those at risk for lung cancer as well as in the tumor microenvironment. EGF significantly enhanced TGF-beta1-mediated induction of COX-2 and corresponding prostaglandin E2 (PGE2) production. TGF-beta1 and EGF induced COX-2 at the transcriptional and post-transcriptional levels. EGF receptor (EGFR) inhibition, neutralizing antibody against amphiregulin, or mitogen-activated protein kinase kinase (MEK) inhibition blocked TGF-beta1-mediated COX-2 induction. COX-2 induction by TGF-beta1 depended upon Smad3 signaling and required the activity of EGFR or its downstream mediators. Autocrine amphiregulin signaling maintains EGFR in a constitutively active state in HBECs, allowing for COX-2 induction by TGF-beta1. Thus, EGFR ligands, which are abundant in the pulmonary microenvironment of those at risk for lung cancer, potentiate and are required for COX-2 induction by TGF-beta1 in HBEC. These findings emphasize the central role of EGFR signaling in COX-2 induction by TGF-beta1 and suggest that inhibition of EGFR signaling should be investigated further for lung cancer prevention.


Subject(s)
Bronchi/cytology , Cyclooxygenase 2/biosynthesis , ErbB Receptors/physiology , Respiratory Mucosa/enzymology , Signal Transduction/physiology , Transforming Growth Factor beta1/physiology , Bronchi/enzymology , Cell Line, Transformed , Dinoprostone/biosynthesis , Enzyme Induction/physiology , Humans , Respiratory Mucosa/cytology
7.
Mol Carcinog ; 46(11): 887-900, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17477362

ABSTRACT

Our previous proteomic investigation of lung neoplasia in vitro demonstrated a high concentration of the lung cancer biomarker and splicing factor, hnRNP A2/B1, in the transformed mouse lung epithelial cell line, E9. Since changes in pre-mRNA splicing profoundly affect neoplastic progression, we examined hnRNP A2/B1 expression in chemically induced primary mouse lung tumors, an in vivo model of pulmonary adencocarcinoma. Tumor hnRNP A2/B1 content and spatial distribution assessed by immunohistochemistry varied with stage of progression, genetic background, and whether tumors were induced by a single agent (urethane) or by 2-stage initiation/promotion (3-methylcholanthrene/butylated hydroxytoluene) carcinogenesis. To address mechanisms governing hnRNP A2/B1 expression changes, we utilized in vitro models. hnRNP A2/B1 protein was overexpressed in E9, the spontaneous tranformant of immortalized but non-neoplastic E10 cells, but expression was not strictly a function of enhanced proliferative rate in neoplastic cells. Elevated mRNA content was positively associated with cell division in both E10 and E9, but hnRNP A2/B1 protein levels decreased in proliferating E10 cells. The increased mRNA reflected enhanced mRNA stability, as shown by measuring time-dependent mRNA decay after inhibiting transcription. Dysregulation of hnRNP A2/B1 expression during lung neoplasia in vivo thus depends on complex gene-environmental interactions that affect cell type-specific changes in mRNA processing and, most probably, the rates of translation and/or protein degradation.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Lung Neoplasms/genetics , Animals , Biomarkers, Tumor , Butylated Hydroxytoluene , Carcinogens , Cell Line , Cell Proliferation , Cell Transformation, Neoplastic , Half-Life , Lung/cytology , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Male , Methylcholanthrene , Mice , Mice, Inbred BALB C , RNA, Messenger/metabolism , Species Specificity , Up-Regulation , Urethane
8.
Carcinogenesis ; 24(4): 651-7, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12727792

ABSTRACT

In order to examine how tumorigenicity is abrogated by gap junctional intercellular communication (GJIC), protein expression was analyzed in four related mouse lung epithelial cell lines that vary in their GJIC status and neoplastic potential. Since alterations in protein expression underlie neoplastic behavior, this proteomic analysis provides insights into the molecular pathogenesis of lung cancer. E10, an immortalized but non-tumorigenic cell line derived from alveolar type II pneumocytes, has functional GJIC. E9, a spontaneous transformant of E10, is GJIC-deficient and is tumorigenic upon injection into a syngeneic mouse. Stable transfection of E9 with Gja1, the gene for the gap junctional protein, connexin 43, re-established GJIC and rendered this line (designated E9-2) non-tumorigenic; the vector transfection control line, E9-41, remains tumorigenic. Proteins extracted from these cell lines were separated by two-dimensional electrophoresis (2DE) and visualized by Coomassie blue staining. We consistently observed differential expression of 27 proteins between E10 and E9 and identified 11 of these by peptide mass mapping. The functions of these proteins include stress response, cytoskeletal structure, signal transduction, apoptosis, immune response, pre-mRNA processing, and carbohydrate metabolism. Gja1 transfection affected the concentrations of four of these proteins, viz. PDI, alpha-enolase, aldolase A, and gelsolin-like protein. PDI concentration was most profoundly affected; E10 cells contain twice as much PDI as E9, and PDI was restored to E10-like levels in the E9-2 transfectant line while remaining at E9-like levels in the vector control E9-41 cells. An association between connexin 43 and PDI expression was also observed in a second set of independently derived type II cell lines. The PDI superfamily has multiple cellular roles including chaperoning assembled glycoproteins, regulating the activities of transcription factors, and regulating disulfide bond formation.


Subject(s)
Connexin 43/genetics , Gap Junctions/genetics , Genes , Lung Neoplasms/genetics , Proteomics , Animals , Epithelial Cells/pathology , Fructose-Bisphosphate Aldolase/metabolism , Gelsolin/metabolism , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Phosphopyruvate Hydratase/metabolism , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...