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










Publication year range
1.
Behav Brain Res ; 467: 114991, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38614209

ABSTRACT

Stroke is a leading cause of death and disability in the United States. Most strokes are ischemic, resulting in both cognitive and motor impairments. Animal models of ischemic stroke such as the distal middle cerebral artery occlusion (dMCAO) and photothrombotic stroke (PTS) procedures have become invaluable tools, with their own advantages and disadvantages. The dMCAO model is clinically relevant as it occludes the artery most affected in humans, but yields variability in the infarct location as well as the behavioral and cognitive phenotypes disrupted. The PTS model has the advantage of allowing for targeted location of infarct, but is less clinically relevant. The present study evaluates phenotype disruption over time in mice subjected to either dMCAO, PTS, or a sham surgery. Post-surgery, animals were tested over 28 days on standard motor tasks (grid walk, cylinder, tapered beam, and rotating beam), as well as a novel odor-based operant task; the 5:1 Odor Discrimination Task (ODT). Results demonstrate a significantly greater disturbance of motor control with PTS as compared with Sham and dMCAO. Disruption of the PTS group was detected up to 28 days post-stroke on the grid walk, and up to 7 days on the rotating and tapered beam tasks. PTS also led to significant short-term disruption of ODT performance (1-day post-surgery), exclusively in males, which appeared to be driven by motoric disruption of the lick response. Together, this data provides critical insights into the selection and optimization of animal models for ischemic stroke research. Notably, the PTS procedure was best suited for producing disruptions of motor behavior that can be detected with common behavioral assays and are relatively enduring, as is observed in human stroke.


Subject(s)
Disease Models, Animal , Infarction, Middle Cerebral Artery , Mice, Inbred C57BL , Animals , Male , Infarction, Middle Cerebral Artery/physiopathology , Infarction, Middle Cerebral Artery/complications , Mice , Stroke/physiopathology , Stroke/complications , Motor Activity/physiology , Thrombotic Stroke , Female , Odorants , Discrimination, Psychological/physiology , Behavior, Animal/physiology , Ischemic Stroke/physiopathology
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.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
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
15.
Am J Respir Crit Care Med ; 177(9): 959-69, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18263801

ABSTRACT

RATIONALE: Nuclear factor (NF)-kappaB is a prominent proinflammatory transcription factor that plays a critical role in allergic airway disease. Previous studies demonstrated that inhibition of NF-kappaB in airway epithelium causes attenuation of allergic inflammation. OBJECTIVES: We sought to determine if selective activation of NF-kappaB within the airway epithelium in the absence of other agonists is sufficient to cause allergic airway disease. METHODS: A transgenic mouse expressing a doxycycline (Dox)-inducible, constitutively active (CA) version of inhibitor of kappaB (IkappaB) kinase-beta (IKKbeta) under transcriptional control of the rat CC10 promoter, was generated. MEASUREMENTS AND MAIN RESULTS: After administration of Dox, expression of the CA-IKKbeta transgene induced the nuclear translocation of RelA in airway epithelium. IKKbeta-triggered activation of NF-kappaB led to an increased content of neutrophils and lymphocytes, and concomitant production of proinflammatory mediators, responses that were not observed in transgenic mice not receiving Dox, or in transgene-negative littermate control animals fed Dox. Unexpectedly, expression of the IKKbeta transgene in airway epithelium was sufficient to cause airway hyperresponsiveness and smooth muscle thickening in absence of an antigen sensitization and challenge regimen, the presence of eosinophils, or the induction of mucus metaplasia. CONCLUSIONS: These findings demonstrate that selective activation NF-kappaB in airway epithelium is sufficient to induce airway hyperresponsiveness and smooth muscle thickening, which are both critical features of allergic airway disease.


Subject(s)
Bronchial Hyperreactivity/metabolism , Bronchitis/metabolism , Doxycycline/pharmacology , NF-kappa B/metabolism , Respiratory Mucosa/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Bronchial Hyperreactivity/pathology , Bronchitis/pathology , Cells, Cultured , Disease Models, Animal , Furans , Gene Expression , I-kappa B Kinase/drug effects , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Lymphocytes/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth/pathology , NF-kappa B/drug effects , Neutrophils/pathology , RNA/genetics , Respiratory Mucosa/pathology , Thiophenes , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...