Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Immunol ; 204(4): 763-774, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31924651

ABSTRACT

Asthma is a chronic disorder characterized by inflammation, mucus metaplasia, airway remodeling, and hyperresponsiveness. We recently showed that IL-1-induced glycolytic reprogramming contributes to allergic airway disease using a murine house dust mite model. Moreover, levels of pyruvate kinase M2 (PKM2) were increased in this model as well as in nasal epithelial cells from asthmatics as compared with healthy controls. Although the tetramer form of PKM2 converts phosphoenolpyruvate to pyruvate, the dimeric form of PKM2 has alternative, nonglycolysis functions as a transcriptional coactivator to enhance the transcription of several proinflammatory cytokines. In the current study, we examined the impact of PKM2 on the pathogenesis of house dust mite-induced allergic airways disease in C57BL/6NJ mice. We report, in this study, that activation of PKM2, using the small molecule activator, TEPP46, augmented PKM activity in lung tissues and attenuated airway eosinophils, mucus metaplasia, and subepithelial collagen. TEPP46 attenuated IL-1ß-mediated airway inflammation and expression of proinflammatory mediators. Exposure to TEPP46 strongly decreased the IL-1ß-mediated increases in thymic stromal lymphopoietin (TSLP) and GM-CSF in primary tracheal epithelial cells isolated from C57BL/6NJ mice. We also demonstrate that IL-1ß-mediated increases in nuclear phospho-STAT3 were decreased by TEPP46. Finally, STAT3 inhibition attenuated the IL-1ß-induced release of TSLP and GM-CSF, suggesting that the ability of PKM2 to phosphorylate STAT3 contributes to its proinflammatory function. Collectively, these results demonstrate that the glycolysis-inactive form of PKM2 plays a crucial role in the pathogenesis of allergic airways disease by increasing IL-1ß-induced proinflammatory signaling, in part, through phosphorylation of STAT3.


Subject(s)
Asthma/immunology , Hypersensitivity/immunology , Pneumonia/immunology , Pyruvate Kinase/immunology , Signal Transduction/immunology , Airway Remodeling/physiology , Animals , Asthma/metabolism , Female , Hypersensitivity/metabolism , Male , Mice , Mice, Inbred C57BL , Pneumonia/metabolism , Pyroglyphidae/immunology , Pyruvate Kinase/metabolism
2.
Oncotarget ; 9(1): 293-305, 2018 Jan 02.
Article in English | MEDLINE | ID: mdl-29416614

ABSTRACT

Malignant mesothelioma is an aggressive cancer in desperate need of treatment. We have previously shown that extracellular signaling regulated kinase 5 (ERK5) plays an important role in mesothelioma pathogenesis using ERK5 silenced human mesothelioma cells exhibiting significantly reduced tumor growth in immunocompromised mice. Here, we used a specific ERK 5 inhibitor, XMD8-92 in various in vitro and in vivo models to demonstrate that inhibition of ERK5 can slow down mesothelioma tumorigenesis. First, we show a dose dependent toxicity of XMD8-92 to 2 human mesothelioma cell lines growing as a monolayer. We also demonstrate the inhibition of ERK5 phosphorylation in various human mesothelioma cell lines by XMD8-92. We further confirmed the toxicity of XMD8-92 towards mesothelioma cell lines grown as spheroids in a 3-D model as well as in intraperitoneal (immune-competent) and intrapleural (immune-deficient) mouse models with and without chemotherapeutic drugs. To ascertain the mechanism, we explored the role of the nod-like receptor family member containing a pyrin domain 3 (NLRP3) inflammasome in the process. We found XMD8-92 attenuated naïve and chemotherapeutic-induced inflammasome priming and activation in mesothelioma cells. It can thus be concluded that ERK5 inhibition attenuates mesothelioma tumor growth and this phenomenon in part is regulated by the inflammasome.

3.
Am J Pathol ; 187(3): 665-678, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28056339

ABSTRACT

Despite the causal relationship established between malignant mesothelioma (MM) and asbestos exposure, the exact mechanism by which asbestos induces this neoplasm and other asbestos-related diseases is still not well understood. MM is characterized by chronic inflammation, which is believed to play an intrinsic role in the origin of this disease. We recently found that asbestos activates the nod-like receptor family member containing a pyrin domain 3 (NLRP3) inflammasome in a protracted manner, leading to an up-regulation of IL-1ß and IL-18 production in human mesothelial cells. Combined with biopersistence of asbestos fibers, we hypothesize that this creates an environment of chronic IL-1ß signaling in human mesothelial cells, which may promote mesothelial to fibroblastic transition (MFT) in an NLRP3-dependent manner. Using a series of experiments, we found that asbestos induces a fibroblastic transition of mesothelial cells with a gain of mesenchymal markers (vimentin and N-cadherin), whereas epithelial markers, such as E-cadherin, are down-regulated. Use of siRNA against NLRP3, recombinant IL-1ß, and IL-1 receptor antagonist confirmed the role of NLRP3 inflammasome-dependent IL-1ß in the process. In vivo studies using wild-type and various inflammasome component knockout mice also revealed the process of asbestos-induced mesothelial to fibroblastic transition and its amelioration in caspase-1 knockout mice. Taken together, our data are the first to suggest that asbestos induces mesothelial to fibroblastic transition in an inflammasome-dependent manner.


Subject(s)
Asbestos/adverse effects , Epithelium/pathology , Fibroblasts/pathology , Inflammasomes/metabolism , Animals , Biomarkers/metabolism , Caspase 1/metabolism , Cell Line , Cell Shape/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelium/metabolism , Fibroblasts/metabolism , Gene Expression Profiling , Gene Expression Regulation , Humans , Interleukin-1beta/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Peritoneum/metabolism , Peritoneum/pathology , Signal Transduction/genetics
4.
Cancer Nanotechnol ; 6: 1, 2015.
Article in English | MEDLINE | ID: mdl-25642297

ABSTRACT

INTRODUCTION: Five year survival for metastatic melanoma (MM) is very low at <10%. Therapeutic options have been limited secondary to systemic toxicity. As a result there has been a growing movement towards developing targeted drug delivery models. Prior research of this group has demonstrated the effectiveness of acid-prepared mesoporous spheres (APMS-TEG) in delivering chemotherapeutic agents at a lower effective dose than systemic administration. This study aims to assess the ability of the previously developed APMS-TEG particles to deliver therapeutic doses of docetaxel for the treatment of melanoma. METHODS: In vitro experiments were performed to assess docetaxel loading onto APMS-TEG particles and release kinetics. Toxicity experiments were performed using docetaxel and docetaxel loaded APMS-TEG. The effect on cell growth was assessed using the MelJuSo, UACC903, and WM1205 melanoma cell lines. RESULTS: Docetaxel demonstrated statistically significant dose dependent reduction in growth of melanoma cells. In all three cell lines, doses of 1 nM were sufficient to produce statistically significant reduction in cell growth. Scanning electron micrographs demonstrate increased uptake of APMS-TEG particles by melanoma cells in the first 24 hours, with the majority within the first 4 hours. Unloaded APMS particles had no effect on the melanoma cells, demonstrating that the particles themselves are not toxic. APMS-TEG particles had a peak release of drug within the first hour, with equilibration thereafter. The 5, 10, and 20 nM loaded particles all had statistically significant reduction in cell growth than the control groups. DISCUSSION: The high potency against melanoma cells makes docetaxel a suitable choice for loading into APMS-TEG particles. Docetaxel loaded APMS-TEG particles demonstrate significant activity against malignant melanoma and thus offer an innovative approach to the treatment of metastatic melanoma.

5.
Am J Pathol ; 184(10): 2816-27, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25111229

ABSTRACT

Malignant mesothelioma (MM) is an aggressive tumor with no treatment regimen. Previously we have demonstrated that cyclic AMP response element binding protein (CREB) is constitutively activated in MM tumor cells and tissues and plays an important role in MM pathogenesis. To understand the role of CREB in MM tumor growth, we generated CREB-inhibited MM cell lines and performed in vitro and in vivo experiments. In vitro experiments demonstrated that CREB inhibition results in significant attenuation of proliferation and drug resistance of MM cells. CREB-silenced MM cells were then injected into severe combined immunodeficiency mice, and tumor growth in s.c. and i.p. models of MM was followed. We observed significant inhibition in MM tumor growth in both s.c. and i.p. models and the presence of a chemotherapeutic drug, doxorubicin, further inhibited MM tumor growth in the i.p. model. Peritoneal lavage fluids from CREB-inhibited tumor-bearing mice showed a significantly reduced total cell number, differential cell counts, and pro-inflammatory cytokines and chemokines (IL-6, IL-8, regulated on activation normal T cell expressed and secreted, monocyte chemotactic protein-1, and vascular endothelial growth factor). In vitro studies showed that asbestos-induced inflammasome/inflammation activation in mesothelial cells was CREB dependent, further supporting the role of CREB in inflammation-induced MM pathogenesis. In conclusion, our data demonstrate the involvement of CREB in the regulation of MM pathogenesis by regulation of inflammation.


Subject(s)
CREB-Binding Protein/metabolism , Lung Neoplasms/pathology , Mesothelioma/pathology , Animals , Asbestos/adverse effects , Cell Line, Tumor , Chemokine CCL2/metabolism , Chemokines/metabolism , Disease Models, Animal , Doxorubicin/pharmacology , Gene Expression Profiling , Heterografts , Humans , Inflammation , Interleukin-6/metabolism , Interleukin-8/metabolism , Lung Neoplasms/metabolism , Male , Mesothelioma/metabolism , Mesothelioma, Malignant , Mice , Mice, SCID , Oligonucleotide Array Sequence Analysis , Phosphorylation , Vascular Endothelial Growth Factor A/metabolism
6.
Part Fibre Toxicol ; 11: 24, 2014 May 20.
Article in English | MEDLINE | ID: mdl-24885895

ABSTRACT

BACKGROUND: Asbestos exposure is related to various diseases including asbestosis and malignant mesothelioma (MM). Among the pathogenic mechanisms proposed by which asbestos can cause diseases involving epithelial and mesothelial cells, the most widely accepted one is the generation of reactive oxygen species and/or depletion of antioxidants like glutathione. It has also been demonstrated that asbestos can induce inflammation, perhaps due to activation of inflammasomes. METHODS: The oxidation state of thioredoxin was analyzed by redox Western blot analysis and ROS generation was assessed spectrophotometrically as a read-out of solubilized formazan produced by the reduction of nitrotetrazolium blue (NTB) by superoxide. Quantitative real time PCR was used to assess changes in gene transcription. RESULTS: Here we demonstrate that crocidolite asbestos fibers oxidize the pool of the antioxidant, Thioredoxin-1 (Trx1), which results in release of Thioredoxin Interacting Protein (TXNIP) and subsequent activation of inflammasomes in human mesothelial cells. Exposure to crocidolite asbestos resulted in the depletion of reduced Trx1 in human peritoneal mesothelial (LP9/hTERT) cells. Pretreatment with the antioxidant dehydroascorbic acid (a reactive oxygen species (ROS) scavenger) reduced the level of crocidolite asbestos-induced Trx1 oxidation as well as the depletion of reduced Trx1. Increasing Trx1 expression levels using a Trx1 over-expression vector, reduced the extent of Trx1 oxidation and generation of ROS by crocidolite asbestos, and increased cell survival. In addition, knockdown of TXNIP expression by siRNA attenuated crocidolite asbestos-induced activation of the inflammasome. CONCLUSION: Our novel findings suggest that extensive Trx1 oxidation and TXNIP dissociation may be one of the mechanisms by which crocidolite asbestos activates the inflammasome and helps in development of MM.


Subject(s)
Asbestos, Crocidolite/toxicity , Inflammation/pathology , Thioredoxins/drug effects , Acetylcysteine/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Blotting, Western , Caspase 1/metabolism , Cell Line, Tumor , Dehydroascorbic Acid/metabolism , Dinitrochlorobenzene/toxicity , Enzyme Activation/drug effects , Epithelium/drug effects , Epithelium/pathology , Gene Knockdown Techniques , Humans , L-Lactate Dehydrogenase/metabolism , RNA, Small Interfering , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Thioredoxin Reductase 1/metabolism , Thioredoxins/genetics
7.
Am J Respir Cell Mol Biol ; 51(5): 595-603, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24940987

ABSTRACT

Malignant mesothelioma (MM), lung cancers, and asbestosis are hyperproliferative diseases associated with exposures to asbestos. All have a poor prognosis; thus, the need to develop novel and effective therapies is urgent. Vandetanib (Van) (ZD6474, ZACTIMA) is a tyrosine kinase inhibitor that has shown equivocal results in clinical trials for advanced non-small cell lung cancer. However, tyrosine kinase inhibitors alone have shown no significant clinical activity in phase II trials of patients with unresectable MM. Using epithelioid (HMESO) and sarcomatoid (H2373) human MM lines, the efficacy of tumor cell killing and signaling pathways modulated by Van with and without doxorubicin (Dox) was examined. Van alone reduced total cell numbers in HMESO MM and synergistically increased the toxicity of Dox in HMESO and H2373 cells. Most importantly, we identified two novel cell survival/resistance pathways, ERK5 and cyclic AMP response element binding protein (CREB), that were inhibited by Van and Dox. After silencing of either ERK5 or CREB, significant decreases in cell numbers in the Dox-resistant sarcomatoid H2373 line were observed. Results suggest that a plethora of cell signaling pathways associated with cell survival are induced by Dox but inhibited by the addition of Van in MM. Data from our study support the combined efficacy of Van and Dox as a novel approach in the treatment of MM that is further enhanced by blocking ERK5 or CREB signaling cascades.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Doxorubicin/pharmacology , MAP Kinase Signaling System/physiology , Mesothelioma/drug therapy , Mitogen-Activated Protein Kinase 7/metabolism , Piperidines/pharmacology , Quinazolines/pharmacology , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cyclic AMP Response Element-Binding Protein/genetics , Doxorubicin/toxicity , Drug Synergism , Humans , MAP Kinase Signaling System/drug effects , Mesothelioma/metabolism , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/genetics , Neoplasms, Connective Tissue/drug therapy , Neoplasms, Connective Tissue/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Piperidines/toxicity , Quinazolines/toxicity , RNA, Small Interfering/genetics , Sarcoma/drug therapy , Sarcoma/metabolism
8.
Cancer Prev Res (Phila) ; 7(3): 330-40, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24431405

ABSTRACT

Inflammation is a key mediator in the development of malignant mesothelioma, which has a dismal prognosis and poor therapeutic strategies. Curcumin, a naturally occurring polyphenol in turmeric, has been shown to possess anticarcinogenic properties through its anti-inflammatory effects. Inflammasomes, a component of inflammation, control the activation of caspase-1 leading to pyroptosis and processing of proinflammatory cytokines, interleukin (IL)-1ß and IL-18. In the present study, we investigate the role of curcumin in pyroptotic cell death of malignant mesothelioma cells. Using in vitro models with mouse and human malignant mesothelioma cells, curcumin is shown to induce pyroptosis through activation of caspase-1 and increased release of high-mobility group box 1 (HMGB1) without processing of IL-1ß and IL-18. Absence of IL-1ß processing in response to curcumin-mediated caspase-1 activation is attributed to blockade of pro-IL-1ß priming through inhibition of the NF-κB pathway. Furthermore, curcumin's cytotoxicity in malignant mesothelioma cells is demonstrated to be dependent on pyroptosis as inhibition of caspase-1 resulted in protection against curcumin-induced cell death. We also demonstrate that curcumin-mediated caspase-1 activation is oxidant dependent by using N-acetyl-L-cysteine (NAC) to inhibit pyroptosis. PCR array analysis using the human inflammasome template revealed that curcumin significantly downregulated levels of inflammasome-related gene expression involved in inflammation, e.g., NF-κB, toll-like receptors (TLR), and IL-1ß. Our data indicate that curcumin has a double effect on malignant mesothelioma cells through induction of pyroptosis while subsequently protecting against inflammation.


Subject(s)
Apoptosis/drug effects , Curcumin/pharmacology , Inflammasomes/drug effects , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Mesothelioma/immunology , Mesothelioma/pathology , Animals , Apoptosis/genetics , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 1/metabolism , Cytokines/metabolism , Humans , Inflammasomes/genetics , Inflammasomes/metabolism , Lung Neoplasms/metabolism , Mesothelioma/metabolism , Mesothelioma, Malignant , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , RNA, Small Interfering/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured
9.
BMC Cancer ; 13: 400, 2013 Sep 11.
Article in English | MEDLINE | ID: mdl-24024776

ABSTRACT

BACKGROUND: Malignant mesotheliomas (MMs) are chemoresistant tumors related to exposure to asbestos fibers. The long latency period of MM (30-40 yrs) and heterogeneity of tumor presentation make MM difficult to diagnose and treat at early stages. Currently approved second-line treatments following surgical resection of MMs include a combination of cisplatin or carboplatin (delivered systemically) and pemetrexed, a folate inhibitor, with or without subsequent radiation. The systemic toxicities of these treatments emphasize the need for more effective, localized treatment regimens. METHODS: Acid-prepared mesoporous silica (APMS) microparticles were loaded with doxorubicin (DOX) and modified externally with a mesothelin (MB) specific antibody before repeated intraperitoneal (IP) injections into a mouse xenograft model of human peritoneal MM. The health/weight of mice, tumor volume/weight, tumor necrosis and cell proliferation were evaluated in tumor-bearing mice receiving saline, DOX high (0.2 mg/kg), DOX low (0.05 mg/kg), APMS-MB, or APMS-MB-DOX (0.05 mg/kg) in saline. RESULTS: Targeted therapy (APMS-MB-DOX at 0.05 mg/kg) was more effective than DOX low (0.05 mg/kg) and less toxic than treatment with DOX high (0.2 mg/kg). It also resulted in the reduction of tumor volume without loss of animal health and weight, and significantly decreased tumor cell proliferation. High pressure liquid chromatography (HPLC) of tumor tissue confirmed that APMS-MB-DOX particles delivered DOX to target tissue. CONCLUSIONS: Data suggest that targeted therapy results in greater chemotherapeutic efficacy with fewer adverse side effects than administration of DOX alone. Targeted microparticles are an attractive option for localized drug delivery.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Doxorubicin/administration & dosage , Drug Delivery Systems , GPI-Linked Proteins/antagonists & inhibitors , Mesothelioma/metabolism , Mesothelioma/pathology , Microspheres , Animals , Body Weight , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , GPI-Linked Proteins/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology , Injections, Intraperitoneal , Ki-67 Antigen/metabolism , Macrophages/pathology , Mesothelin , Mesothelioma/drug therapy , Mice , Necrosis/drug therapy , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
10.
Part Fibre Toxicol ; 10: 39, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23937860

ABSTRACT

BACKGROUND: Pleural fibrosis and malignant mesotheliomas (MM) occur after exposures to pathogenic fibers, yet the mechanisms initiating these diseases are unclear. RESULTS: We document priming and activation of the NLRP3 inflammasome in human mesothelial cells by asbestos and erionite that is causally related to release of IL-1ß, IL-6, IL-8, and Vascular Endothelial Growth Factor (VEGF). Transcription and release of these proteins are inhibited in vitro using Anakinra, an IL-1 receptor antagonist that reduces these cytokines in a human peritoneal MM mouse xenograft model. CONCLUSIONS: These novel data show that asbestos-induced priming and activation of the NLRP3 inflammasome triggers an autocrine feedback loop modulated via the IL-1 receptor in mesothelial cell type targeted in pleural infection, fibrosis, and carcinogenesis.


Subject(s)
Asbestos, Crocidolite/toxicity , Autocrine Communication , Carrier Proteins/metabolism , Cytokines/metabolism , Epithelium/drug effects , Inflammasomes/drug effects , Inflammation Mediators/metabolism , Mesothelioma/chemically induced , Zeolites/toxicity , Animals , Cell Line, Tumor , Cytokines/genetics , Dose-Response Relationship, Drug , Epithelium/immunology , Epithelium/pathology , Humans , Inflammasomes/immunology , Interleukin 1 Receptor Antagonist Protein/pharmacology , Mesothelioma/drug therapy , Mesothelioma/genetics , Mesothelioma/immunology , Mesothelioma/pathology , Mice , Mice, SCID , NLR Family, Pyrin Domain-Containing 3 Protein , Primary Cell Culture , RNA, Messenger/metabolism , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/metabolism , Time Factors , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays
11.
Clin Cancer Res ; 19(8): 2071-83, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23446998

ABSTRACT

PURPOSE: Malignant mesothelioma is a devastating disease with a need for new treatment strategies. In the present study, we showed the importance of extracellular signal-regulated kinase 5 (ERK5) in malignant mesothelioma tumor growth and treatment. EXPERIMENTAL DESIGN: ERK5 as a target for malignant mesothelioma therapy was verified using mesothelial and mesothelioma cell lines as well as by xenograft severe combined immunodeficient (SCID) mouse models. RESULTS: We first showed that crocidolite asbestos activated ERK5 in LP9 cells and mesothelioma cell lines exhibit constitutive activation of ERK5. Addition of doxorubicin resulted in further activation of ERK5 in malignant mesothelioma cells. ERK5 silencing increased doxorubicin-induced cell death and doxorubicin retention in malignant mesothelioma cells. In addition, shERK5 malignant mesothelioma lines exhibited both attenuated colony formation on soft agar and invasion of malignant mesothelioma cells in vitro that could be related to modulation of gene expression linked to cell proliferation, apoptosis, migration/invasion, and drug resistance as shown by microarray analysis. Most importantly, injection of shERK5 malignant mesothelioma cell lines into SCID mice showed significant reduction in tumor growth using both subcutaneous and intraperitoneal models. Assessment of selected human cytokine profiles in peritoneal lavage fluid from intraperitoneal shERK5 and control tumor-bearing mice showed that ERK5 was critical in regulation of various proinflammatory (RANTES/CCL5, MCP-1) and angiogenesis-related (interleukin-8, VEGF) cytokines. Finally, use of doxorubicin and cisplatin in combination with ERK5 inhibition showed further reduction in tumor weight and volume in the intraperitoneal model of tumor growth. CONCLUSION: ERK5 inhibition in combination with chemotherapeutic drugs is a beneficial strategy for combination therapy in patients with malignant mesothelioma.


Subject(s)
Mesothelioma/genetics , Mesothelioma/therapy , Mitogen-Activated Protein Kinase 7/genetics , RNA Interference , Animals , Antineoplastic Agents/pharmacology , Asbestos, Crocidolite/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cisplatin/pharmacology , Combined Modality Therapy , Cytokines/genetics , Cytokines/metabolism , Doxorubicin/pharmacology , Enzyme Activation/drug effects , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mesothelioma/pathology , Mice , Mice, SCID , Mitogen-Activated Protein Kinase 7/metabolism , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Tumor Burden/drug effects , Tumor Burden/genetics , Xenograft Model Antitumor Assays
12.
J Histochem Cytochem ; 60(9): 658-74, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22723527

ABSTRACT

Pleural and peritoneal mesotheliomas (MMs) are chemoresistant tumors with no effective therapeutic strategies. The authors first injected multifunctional, acid-prepared mesoporous spheres (APMS), microparticles functionalized with tetraethylene glycol oligomers, intraperitoneally into rodents. Biodistribution of APMS was observed in major organs, peritoneal lavage fluid (PLF), and urine of normal mice and rats. After verification of increased mesothelin in human mesotheliomas injected into severe combined immunodeficient (SCID) mice, APMS were then functionalized with an antibody to mesothelin (APMS-MB) or bovine serum albumin (BSA), a nonspecific protein control, and tumor targeting was evaluated by inductively coupled plasma mass spectrometry and multifluorescence confocal microscopy. Some APMS were initially cleared via the urine over a 24 hr period, and small amounts were observed in liver, spleen, and kidneys at 24 hr and 6 days. Targeting with APMS-MB increased APMS uptake in mesenteric tumors at 6 days. Approximately 10% to 12% of the initially injected amount was observed in both spheroid and mesenteric MM at this time point. The data suggest that localized delivery of APMS-MB into the peritoneal cavity after encapsulation of drugs, DNA, or macromolecules is a novel therapeutic approach for MM and other tumors (ovarian and pancreatic) that overexpress mesothelin.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antigens, Differentiation/metabolism , Drug Carriers/chemistry , GPI-Linked Proteins/metabolism , Mesothelioma/metabolism , Silicon Dioxide/chemistry , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics , Antigens, Differentiation/immunology , Cattle , Cell Line, Tumor , Dogs , Drug Carriers/pharmacokinetics , Fluorescent Dyes/chemistry , GPI-Linked Proteins/immunology , Gadolinium/chemistry , Humans , Mesentery , Mesothelin , Mice , Mice, SCID , Particle Size , Peritoneal Neoplasms/metabolism , Rats , Serum Albumin, Bovine/chemistry , Silicon Dioxide/pharmacokinetics , Spheroids, Cellular/metabolism , Tissue Distribution , Transplantation, Heterologous
13.
Int J Cancer ; 129(5): 1075-86, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21710492

ABSTRACT

Members of the extracellular signal-regulated kinase (ERK) family may have distinct roles in the development of cell injury and repair, differentiation and carcinogenesis. Here, we show, using a synthetic small-molecule MEK1/2 inhibitor (U0126) and RNA silencing of ERK1 and 2, comparatively, that ERK2 is critical to transformation and homeostasis of human epithelioid malignant mesotheliomas (MMs), asbestos-induced tumors with a poor prognosis. Although MM cell (HMESO) lines stably transfected with shERK1 or shERK2 both exhibited significant decreases in cell proliferation in vitro, injection of shERK2 cells, and not shERK1 cells, into immunocompromised severe combined immunodeficiency (SCID) mice showed significant attenuated tumor growth in comparison to shControl (shCon) cells. Inhibition of migration, invasion and colony formation occurred in shERK2 MM cells in vitro, suggesting multiple roles of ERK2 in neoplasia. Microarray and quantitative real-time PCR analyses revealed gene expression that was significantly increased (CASP1, TRAF1 and FAS) or decreased (SEMA3E, RPS6KA2, EGF and BCL2L1) in shERK2-transfected MM cells in contrast to shCon-transfected MM cells. Most striking decreases were observed in mRNA levels of Semaphorin 3 (SEMA3E), a candidate tumor suppressor gene linked to inhibition of angiogenesis. These studies demonstrate a key role of ERK2 in novel gene expression critical to the development of epithelioid MMs. After injection of sarcomatoid human MM (PPMMill) cells into SCID mice, both shERK1 and shERK2 lines showed significant decreased tumor growth, suggesting heterogeneous effects of ERKs in individual MMs.


Subject(s)
Mesothelioma/metabolism , Mesothelioma/pathology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Animals , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Butadienes/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Mesothelioma/drug therapy , Mice , Mice, SCID , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Nitriles/pharmacology , Oligonucleotide Array Sequence Analysis , Pleural Neoplasms/drug therapy , Pleural Neoplasms/metabolism , Pleural Neoplasms/pathology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays
14.
Am J Respir Cell Mol Biol ; 45(5): 906-14, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21454801

ABSTRACT

We hypothesized that normal human mesothelial cells acquire resistance to asbestos-induced toxicity via induction of one or more epidermal growth factor receptor (EGFR)-linked survival pathways (phosphoinositol-3-kinase/AKT/mammalian target of rapamycin and extracellular signal-regulated kinase [ERK] 1/2) during simian virus 40 (SV40) transformation and carcinogenesis. Both isolated HKNM-2 mesothelial cells and a telomerase-immortalized mesothelial line (LP9/TERT-1) were more sensitive to crocidolite asbestos toxicity than an SV40 Tag-immortalized mesothelial line (MET5A) and malignant mesothelioma cell lines (HMESO and PPM Mill). Whereas increases in phosphorylation of AKT (pAKT) were observed in MET5A cells in response to asbestos, LP9/TERT-1 cells exhibited dose-related decreases in pAKT levels. Pretreatment with an EGFR phosphorylation or mitogen-activated protein kinase kinase 1/2 inhibitor abrogated asbestos-induced phosphorylated ERK (pERK) 1/2 levels in both LP9/TERT-1 and MET5A cells as well as increases in pAKT levels in MET5A cells. Transient transfection of small interfering RNAs targeting ERK1, ERK2, or AKT revealed that ERK1/2 pathways were involved in cell death by asbestos in both cell lines. Asbestos-resistant HMESO or PPM Mill cells with high endogenous levels of ERKs or AKT did not show dose-responsive increases in pERK1/ERK1, pERK2/ERK2, or pAKT/AKT levels by asbestos. However, small hairpin ERK2 stable cell lines created from both malignant mesothelioma lines were more sensitive to asbestos toxicity than shERK1 and shControl lines, and exhibited unique, tumor-specific changes in endogenous cell death-related gene expression. Our results suggest that EGFR phosphorylation is causally linked to pERK and pAKT activation by asbestos in normal and SV40 Tag-immortalized human mesothelial cells. They also indicate that ERK2 plays a role in modulating asbestos toxicity by regulating genes critical to cell injury and survival that are differentially expressed in human mesotheliomas.


Subject(s)
Asbestos, Crocidolite/toxicity , Mesothelioma/chemically induced , Mesothelioma/enzymology , Mitogen-Activated Protein Kinase 1/metabolism , Pleural Neoplasms/chemically induced , Pleural Neoplasms/enzymology , Cell Line , Cell Survival/drug effects , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , Gene Expression/drug effects , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , RNA, Small Interfering/metabolism , Signal Transduction/drug effects
15.
Am J Pathol ; 178(5): 1975-85, 2011 May.
Article in English | MEDLINE | ID: mdl-21514415

ABSTRACT

Inflammation and lung remodeling are hallmarks of asbestos-induced fibrosis, but the molecular mechanisms that control these events are unclear. Using laser capture microdissection (LCM) of distal bronchioles in a murine asbestos inhalation model, we show that osteopontin (OPN) is up-regulated by bronchiolar epithelial cells after chrysotile asbestos exposures. In contrast to OPN wild-type mice (OPN(+/+)) inhaling asbestos, OPN null mice (OPN(-/-)) exposed to asbestos showed less eosinophilia in bronchoalveolar lavage fluids, diminished lung inflammation, and decreased mucin production. Bronchoalveolar lavage fluid concentrations of inflammatory cytokines (IL-1ß, IL-4, IL-6, IL-12 subunit p40, MIP1α, MIP1ß, and eotaxin) also were significantly less in asbestos-exposed OPN(-/-) mice. Microarrays performed on lung tissues from asbestos-exposed OPN(+/+) and OPN(-/-) mice showed that OPN modulated the expression of a number of genes (Col1a2, Timp1, Tnc, Eln, and Col3a1) linked to fibrosis via initiation and cross talk between IL-1ß and epidermal growth factor receptor-related signaling pathways. Novel targets of OPN identified include genes involved in cell signaling, immune system/defense, extracellular matrix remodeling, and cell cycle regulation. Although it is unclear whether the present findings are specific to chrysotile asbestos or would be observed after inhalation of other fibers in general, these results highlight new potential mechanisms and therapeutic targets for asbestosis and other diseases (asthma, smoking-related interstitial lung diseases) linked to OPN overexpression.


Subject(s)
Asbestosis/metabolism , Gene Expression Profiling , Inflammation/metabolism , Mucins/biosynthesis , Osteopontin/metabolism , Animals , Asbestos, Serpentine/adverse effects , Asbestosis/genetics , Asbestosis/pathology , Bronchioles/immunology , Bronchioles/metabolism , Bronchioles/pathology , Bronchoalveolar Lavage , Disease Models, Animal , Inflammation/genetics , Inflammation/pathology , Lasers , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microdissection , Osteopontin/genetics , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Up-Regulation
16.
Int J Cancer ; 129(1): 233-44, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-20830711

ABSTRACT

New and effective treatment strategies are desperately needed for malignant mesothelioma (MM), an aggressive cancer with a poor prognosis. We have shown previously that acid-prepared mesoporous microspheres (APMS) are nontoxic after intrapleural or intraperitoneal (IP) administration to rodents. The purpose here was to evaluate the utility of APMS in delivering chemotherapeutic drugs to human MM cells in vitro and in two mouse xenograft models of MM. Uptake and release of doxorubicin (DOX) alone or loaded in APMS (APMS-DOX) were evaluated in MM cells. MM cell death and gene expression linked to DNA damage/repair were also measured in vitro. In two severe combined immunodeficient mouse xenograft models, mice received saline, APMS, DOX or APMS-DOX injected directly into subcutaneous (SC) MM tumors or injected IP after development of human MMs peritoneally. Other mice received DOX intravenously (IV) via tail vein injections. In comparison to DOX alone, APMS-DOX enhanced intracellular uptake of DOX, MM death and expression of GADD34 and TP73. In the SC MM model, 3× weekly SC injections of APMS-DOX or DOX alone significantly inhibited tumor volumes, and systemic DOX administration was lethal. In mice developing IP MMs, significant (p < 0.05) inhibition of mesenteric tumor numbers, weight and volume was achieved using IP administration of APMS-DOX at one-third the DOX concentration required after IP injections of DOX alone. These results suggest APMS are efficacious for the localized delivery of lower effective DOX concentrations in MM and represent a novel means of treating intracavitary tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Doxorubicin/therapeutic use , Mesothelioma/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Chromatography, High Pressure Liquid , Doxorubicin/administration & dosage , Drug Carriers , Humans , Mice , Microscopy, Confocal , Polymerase Chain Reaction
17.
Mol Cancer ; 9: 314, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21159167

ABSTRACT

BACKGROUND: Malignant mesotheliomas (MM) have a poor prognosis, largely because of their chemoresistance to anti-cancer drugs such as doxorubicin (Dox). Here we show using human MM lines that Dox activates extracellular signal-regulated kinases (ERK1 and 2), causally linked to increased expression of ABC transporter genes, decreased accumulation of Dox, and enhanced MM growth. Using the MEK1/2 inhibitor, U0126 and stably transfected shERK1 and shERK2 MM cell lines, we show that inhibition of both ERK1 and 2 sensitizes MM cells to Dox. RESULTS: U0126 significantly modulated endogenous expression of several important drug resistance (BCL2, ABCB1, ABCC3), prosurvival (BCL2), DNA repair (BRCA1, BRCA2), hormone receptor (AR, ESR2, PPARγ) and drug metabolism (CYP3A4) genes newly identified in MM cells. In comparison to shControl lines, MM cell lines stably transfected with shERK1 or shERK2 exhibited significant increases in intracellular accumulation of Dox and decreases in cell viability. Affymetrix microarray analysis on stable shERK1 and shERK2 MM lines showed more than 2-fold inhibition (p ≤ 0.05) of expression of ATP binding cassette genes (ABCG1, ABCA5, ABCA2, MDR/TAP, ABCA1, ABCA8, ABCC2) in comparison to shControl lines. Moreover, injection of human MM lines into SCID mice showed that stable shERK1 or shERK2 lines had significantly slower tumor growth rates in comparison to shControl lines after Dox treatment. CONCLUSIONS: These studies suggest that blocking ERK1 and 2, which play critical roles in multi-drug resistance and survival, may be beneficial in combination with chemotherapeutic drugs in the treatment of MMs and other tumors.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Mesothelioma/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Animals , Antibiotics, Antineoplastic/therapeutic use , Butadienes/pharmacology , Cell Line, Tumor , Doxorubicin/therapeutic use , Flow Cytometry , Humans , Mesothelioma/drug therapy , Mesothelioma/genetics , Mice , Mice, SCID , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Multidrug Resistance-Associated Protein 2 , Nitriles/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays
18.
Part Fibre Toxicol ; 7: 26, 2010 Sep 11.
Article in English | MEDLINE | ID: mdl-20831825

ABSTRACT

BACKGROUND: Exposures to an amphibole fiber in Libby, Montana cause increases in malignant mesothelioma (MM), a tumor of the pleural and peritoneal cavities with a poor prognosis. Affymetrix microarray/GeneSifter analysis was used to determine alterations in gene expression of a human mesothelial cell line (LP9/TERT-1) by a non-toxic concentration (15×10(6) µm2/cm2) of unprocessed Libby six-mix and negative (glass beads) and positive (crocidolite asbestos) controls. Because manganese superoxide dismutase (MnSOD; SOD2) was the only gene upregulated significantly (p < 0.05) at both 8 and 24 h, we measured SOD protein and activity, oxidative stress and glutathione (GSH) levels to better understand oxidative events after exposure to non-toxic (15×10(6) µm2/cm2) and toxic concentrations (75×10(6) µm2/cm2) of Libby six-mix. RESULTS: Exposure to 15×10(6) µm2/cm2 Libby six-mix elicited significant (p < 0.05) upregulation of one gene (SOD2; 4-fold) at 8 h and 111 gene changes at 24 h, including a 5-fold increase in SOD2. Increased levels of SOD2 mRNA at 24 h were also confirmed in HKNM-2 normal human pleural mesothelial cells by qRT-PCR. SOD2 protein levels were increased at toxic concentrations (75×10(6) µm2/cm2) of Libby six-mix at 24 h. In addition, levels of copper-zinc superoxide dismutase (Cu/ZnSOD; SOD1) protein were increased at 24 h in all mineral groups. A dose-related increase in SOD2 activity was observed, although total SOD activity remained unchanged. Dichlorodihydrofluorescein diacetate (DCFDA) fluorescence staining and flow cytometry revealed a dose- and time-dependent increase in reactive oxygen species (ROS) production by LP9/TERT-1 cells exposed to Libby six-mix. Both Libby six-mix and crocidolite asbestos at 75×10(6) µm2/cm2 caused transient decreases (p < 0.05) in GSH for up to 24 h and increases in gene expression of heme oxygenase 1 (HO-1) in LP9/TERT-1 and HKNM-2 cells. CONCLUSIONS: Libby six-mix causes multiple gene expression changes in LP9/TERT-1 human mesothelial cells, as well as increases in SOD2, increased production of oxidants, and transient decreases in intracellular GSH. These events are not observed at equal surface area concentrations of nontoxic glass beads. Results support a mechanistic basis for the importance of SOD2 in proliferation and apoptosis of mesothelial cells and its potential use as a biomarker of early responses to mesotheliomagenic minerals.


Subject(s)
Asbestos, Amphibole/toxicity , Epithelial Cells/drug effects , Oxidative Stress/drug effects , Asbestos, Crocidolite/toxicity , Cells, Cultured , Epithelial Cells/metabolism , Gene Expression/drug effects , Glutathione/metabolism , Heme Oxygenase-1/genetics , Humans , Reactive Oxygen Species/metabolism , Superoxide Dismutase/analysis
19.
ACS Appl Mater Interfaces ; 2(9): 2489-95, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20707315

ABSTRACT

Strategies were developed by which mesoporous microparticles were modified on their external surfaces with tetraethylene glycol (TEG), a protein, or both, leaving the pore surfaces available for modification with a separate moiety, such as a dye. Only particles bifunctionally modified with both TEG and a cell-specific antibody were taken up specifically by a targeted cancer cell line. In contrast to similarly functionalized nanoparticles, endocytosed microparticles were not contained within a lysosome.


Subject(s)
Antibodies, Monoclonal/immunology , Biocompatible Materials/chemistry , Drug Carriers/chemical synthesis , Neoplasms/chemistry , Neoplasms/immunology , Polymers/chemistry , Silicon Dioxide/chemistry , Antibodies, Monoclonal/administration & dosage , Cell Line, Tumor , Humans , Microspheres , Porosity , Surface Properties
20.
Ann N Y Acad Sci ; 1203: 7-14, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20716277

ABSTRACT

Asbestos fibers cause chronic inflammation that may be critical to the development of malignant mesothelioma (MM). Two human MM cell lines (Hmeso, PPM Mill) were used in a SCID mouse xenograft model to assess time-dependent patterns of inflammation and tumor formation. After intraperitoneal (IP) injection of MM cells, mice were euthanized at 7, 14, and 30 days, and peritoneal lavage fluid (PLF) was examined for immune cell profiles and human and mouse cytokines. Increases in human MM-derived IL-6, IL-8, bFGF, and VEGF were observed in mice at 7 days postinjection of either MM line, and a striking neutrophilia was observed at all time points. Free-floating tumor spheroids developed in mice at 14 days, and both spheroids and adherent MM tumor masses occurred in all mice at 30 days. Results suggest that inflammation and cytokine production precede and may be critical to the development of MMs.


Subject(s)
Cell Transformation, Neoplastic/pathology , Inflammation Mediators/physiology , Mesothelioma/pathology , Pleural Neoplasms/pathology , Xenograft Model Antitumor Assays , Animals , Carcinoma/chemistry , Carcinoma/metabolism , Carcinoma/pathology , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic/chemistry , Cell Transformation, Neoplastic/metabolism , Cytokines/biosynthesis , Cytokines/chemistry , Cytokines/physiology , Disease Models, Animal , Fibrosarcoma/chemistry , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Humans , Inflammation Mediators/chemistry , Inflammation Mediators/metabolism , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/physiology , Male , Mesothelioma/chemistry , Mesothelioma/metabolism , Mice , Mice, SCID , Neutrophils/pathology , Pleural Neoplasms/chemistry , Pleural Neoplasms/metabolism , Protein Array Analysis , Time Factors , Xenograft Model Antitumor Assays/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...