Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters











Publication year range
1.
Nat Prod Res ; : 1-9, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39205489

ABSTRACT

The opioid receptors play a pivotal role in the treatment of several neuropsychiatric and neurological disorders. Oroxylum indicum (L.) Kurtz is a very important medicinal plant with several therapeutic applications. It is a main constituent of the Ayurvedic formulation 'Dashmool' used for multifaceted disorders by the Indians. However, the constituents of this plant in neurological conditions have not been well studied. Here, we performed activity-guided isolation of compounds for opioid receptor modulator activity. In the study, we found that the isolated compound baicalein (3) has shown the most potent and competitive antagonistic activity at 20 mg/kg dose in vivo experiments. The acute dose of 3 (20 mg/kg) and pan opioid receptor antagonist naloxone (20 mg/kg) block the morphine-induced antinociception and the paw withdrawal latency decreases up to 8.3 s and 9.6 s, respectively. The in silico studies also support our in vitro data that compound 3 binds with MOR and KOR.

2.
Int J Mol Sci ; 24(10)2023 May 10.
Article in English | MEDLINE | ID: mdl-37239886

ABSTRACT

Adverse lung outcomes from exposure to per-and polyfluoroalkyl substances (PFAS) are known; however, the mechanism of action is poorly understood. To explore this, human bronchial epithelial cells were grown and exposed to varied concentrations of short-chain (perfluorobutanoic acid, perflurobutane sulfonic acid and GenX) or long-chain (PFOA and perfluorooctane sulfonic acid (PFOS)) PFAS, alone or in a mixture to identify cytotoxic concentrations. Non-cytotoxic concentrations of PFAS from this experiment were selected to assess NLRP3 inflammasome activation and priming. We found that PFOA and PFOS alone or in a mixture primed and activated the inflammasome compared with vehicle control. Atomic force microscopy showed that PFOA but not PFOS significantly altered the membrane properties of cells. RNA sequencing was performed on the lungs of mice that had consumed PFOA in drinking water for 14 weeks. Wild type (WT), PPARα knock-out (KO) and humanized PPARα (KI) were exposed to PFOA. We found that multiple inflammation- and immune-related genes were affected. Taken together, our study demonstrated that PFAS exposure could alter lung biology in a significant manner and may contribute to asthma/airway hyper-responsiveness.


Subject(s)
Alkanesulfonic Acids , Environmental Pollutants , Fluorocarbons , Humans , Animals , Mice , Inflammasomes , PPAR alpha , Alkanesulfonic Acids/toxicity , Fluorocarbons/toxicity , Lung/chemistry
3.
Sci Rep ; 13(1): 6052, 2023 04 13.
Article in English | MEDLINE | ID: mdl-37055421

ABSTRACT

The effect of different bioagents such as Trichoderma harzianum, T. viride, T. virens, Pseudomonas fluorescens, and Bacillus subtilis was studied on seed mycoflora, seed germination, root/shoot length, and seedling vigour of cucumber var. Solan Srijan under in vitro conditions. Alternaria sp., Aspergillus sp., and Fusarium spp. were observed on cucumber as seed mycoflora, with T. harzianum showing the greatest inhibition for Alternaria sp. and Fusarium spp., and T. viride showing the greatest inhibition for Aspergillus sp. Cucumber var. Solan Srijan seeds were treated with various bio agents, with T. harzianum being the most effective in increasing seed germination (88.75%), root length (13.58 cm), shoot length (14.58 cm), and seedling vigour (2501.31).


Subject(s)
Cucumis sativus , Seedlings , Germination , Seeds/physiology , Aspergillus
4.
Int J Mol Sci ; 23(23)2022 Dec 06.
Article in English | MEDLINE | ID: mdl-36499762

ABSTRACT

Malignant mesothelioma (MM) is a devastating cancer of mesothelial cells, caused by asbestos exposure. Limited knowledge regarding the detection of asbestos exposure and the early diagnosis of MM, as well as a lack of successful treatment options for this deadly cancer, project an immediate need to understand the mechanism(s) of MM development. With the recent discovery of nano-vesicles, namely exosomes, and their enormous potential to contain signature molecules representative of different diseases, as well as to communicate with distant targets, we were encouraged to explore their role(s) in MM biology. In this review, we summarize what we know so far about exosomes and MM based on our own studies and on published literature from other groups in the field. We expect that the information contained in this review will help advance the field of MM forward by revealing the mechanisms of MM development and survival. Based on this knowledge, future therapeutic strategies for MM can potentially be developed. We also hope that the outcome of our studies presented here may help in the detection of MM.


Subject(s)
Asbestos , Exosomes , Lung Neoplasms , Mesothelioma, Malignant , Mesothelioma , Humans , Mesothelioma/pathology , Lung Neoplasms/pathology , Asbestos/toxicity , Exosomes/pathology
5.
Sci Rep ; 9(1): 11688, 2019 08 12.
Article in English | MEDLINE | ID: mdl-31406207

ABSTRACT

Malignant mesothelioma (MM) is an asbestos-induced cancer arising on the mesothelial surface of organ cavities. MM is essentially incurable without a means of early diagnosis and no successful standard of care. These facts indicate a deep chasm of knowledge that needs to be filled. Our group recently delved into MM tumor biology from the perspective of exosome-contained microRNAs (miRNAs). We discovered that the most abundant miRNAs in MM cancer exosomes were tumor suppressors, particularly miR-16-5p. This observation lead us to hypothesize that MM cells preferentially secreted tumor-suppressor miRNAs via exosomes. Through separate avenues of potential therapeutic advance, we embarked on an innovative strategy to kill MM tumor cells. We employed small molecule inhibitors to block exosome secretion, thereby reducing miR-16-5p exosome loss and replenishing cellular miR-16-5p leading to reduced tumorigenic capacity and miR-16-5p target oncoproteins CCND1 and BCL2. Additionally, we force-fed MM tumor exosomes back to MM tumor cells, which led to cell death, and a reduction in the same oncoproteins. We recapitulated these results with direct transfection of miR-16-5p, confirmed that this is a cancer-cell specific effect, and elucidated a part of the miR-16-5p mechanism of exosome loading.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin D1/genetics , Exosomes/chemistry , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Aniline Compounds/pharmacology , Benzylidene Compounds/pharmacology , Cell Death , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/metabolism , Exosomes/metabolism , Humans , Indoles/pharmacology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Maleimides/pharmacology , Mesothelioma/genetics , Mesothelioma/metabolism , Mesothelioma/pathology , Mesothelioma, Malignant , MicroRNAs/metabolism , Molecular Targeted Therapy/methods , Ornithine/analogs & derivatives , Ornithine/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Small Molecule Libraries/pharmacology , Transfection
6.
Antioxid Redox Signal ; 31(14): 1070-1091, 2019 11 10.
Article in English | MEDLINE | ID: mdl-30799628

ABSTRACT

Significance: The lung is a unique organ, as it is constantly exposed to air, and thus it requires a robust antioxidant defense system to prevent the potential damage from exposure to an array of environmental insults, including oxidants. The peroxiredoxin (PRDX) family plays an important role in scavenging peroxides and is critical to the cellular antioxidant defense system. Recent Advances: Exciting discoveries have been made to highlight the key features of PRDXs that regulate the redox tone. PRDXs do not act in isolation as they require the thioredoxin/thioredoxin reductase/NADPH, sulfiredoxin (SRXN1) redox system, and in some cases glutaredoxin/glutathione, for their reduction. Furthermore, the chaperone function of PRDXs, controlled by the oxidation state, demonstrates the versatility in redox regulation and control of cellular biology exerted by this class of proteins. Critical Issues: Despite the long-known observations that redox perturbations accompany a number of pulmonary diseases, surprisingly little is known about the role of PRDXs in the etiology of these diseases. In this perspective, we review the studies that have been conducted thus far to address the roles of PRDXs in lung disease, or experimental models used to study these diseases. Intriguing findings, such as the secretion of PRDXs and the formation of autoantibodies, raise a number of questions about the pathways that regulate secretion, redox status, and immune response to PRDXs. Future Directions: Further understanding of the mechanisms by which individual PRDXs control lung inflammation, injury, repair, chronic remodeling, and cancer, and the importance of PRDX oxidation state, configuration, and client proteins that govern these processes is needed.


Subject(s)
Lung Diseases/metabolism , Lung/metabolism , Peroxiredoxins/metabolism , Animals , Humans , Oxidation-Reduction
7.
J Cell Biochem ; 119(7): 6266-6273, 2018 07.
Article in English | MEDLINE | ID: mdl-29663493

ABSTRACT

Asbestos-induced diseases like fibrosis and mesothelioma are very aggressive, without any treatment options. These diseases are diagnosed only at the terminal stages due to lack of early stage biomarkers. The recent discovery of exosomes as circulating biomarkers led us to look for exosomal biomarkers of asbestos exposure in mouse blood. In our model, mice were exposed to asbestos as a single bolus dose by oropharyngeal aspiration. Fifty-six days later blood was collected, exosomes were isolated from plasma and characterized and subjected to proteomic analysis using Tandem Mass Tag labeling. We identified many proteins, some of which were more abundant in asbestos exposed mouse serum exosomes, and three selected proteins were validated by immunoblotting. Our study is the first to show that serum exosomal proteomic signatures can reveal some important proteins relevant to asbestos exposure that have the potential to be validated as candidate biomarkers. We hope to extrapolate the positive findings of this study to humans in future studies.


Subject(s)
Asbestos/toxicity , Blood Proteins/metabolism , Carcinogens/toxicity , Exosomes/metabolism , Administration, Oral , Animals , Asbestos/administration & dosage , Blood Proteins/drug effects , Carcinogens/administration & dosage , Exosomes/drug effects , Mice , Mice, Inbred C57BL , Proteomics , Respiratory Aspiration
8.
FASEB J ; 32(8): 4328-4342, 2018 08.
Article in English | MEDLINE | ID: mdl-29553831

ABSTRACT

Asbestos exposure is a determinate cause of many diseases, such as mesothelioma, fibrosis, and lung cancer, and poses a major human health hazard. At this time, there are no identified biomarkers to demarcate asbestos exposure before the presentation of disease and symptoms, and there is only limited understanding of the underlying biology that governs asbestos-induced disease. In our study, we used exosomes, 30-140 nm extracellular vesicles, to gain insight into these knowledge gaps. As inhaled asbestos is first encountered by lung epithelial cells and macrophages, we hypothesize that asbestos-exposed cells secrete exosomes with signature proteomic cargo that can alter the gene expression of mesothelial cells, contributing to disease outcomes like mesothelioma. In the present study using lung epithelial cells (BEAS2B) and macrophages (THP-1), we first show that asbestos exposure causes changes in abundance of some proteins in the exosomes secreted from these cells. Furthermore, exposure of human mesothelial cells (HPM3) to these exosomes resulted in gene expression changes related to epithelial-to-mesenchymal transition and other cancer-related genes. This is the first report to indicate that asbestos-exposed cells secrete exosomes with differentially abundant proteins and that those exosomes have a gene-altering effect on mesothelial cells.-Munson, P., Lam, Y.-W., Dragon, J. MacPherson, M., Shukla, A. Exosomes from asbestos-exposed cells modulate gene expression in mesothelial cells.


Subject(s)
Asbestos/toxicity , Epithelial Cells/physiology , Epithelium/physiology , Exosomes/genetics , Gene Expression/genetics , Lung/physiology , Carcinogens/toxicity , Cell Line , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/physiology , Epithelium/drug effects , Gene Expression/drug effects , Humans , Inhalation Exposure/adverse effects , Lung/drug effects , Macrophages/drug effects , Macrophages/physiology
9.
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.

10.
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
11.
Histochem Cell Biol ; 147(5): 595-604, 2017 May.
Article in English | MEDLINE | ID: mdl-28013367

ABSTRACT

Asbestos exposure leads to malignant mesothelioma (MM), a deadly neoplasm of mesothelial cells of various locations. Although there is no doubt about the role of asbestos in MM tumorigenesis, mechanisms are still not well explored. Recently, our group demonstrated that asbestos causes inflammasome priming and activation in mesothelial cells, which in part is dependent on oxidative stress. Our current study sheds light on yet another mechanism of inflammasome activation by asbestos. Here we show the role of actin polymerization in asbestos-induced activation of the nod-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome. Using human mesothelial cells, we first demonstrate that asbestos and carbon nanotubes induced caspase-1 activation and high-mobility group box 1, interleukin 1 beta and interleukin 18 secretion was blocked by Cytochalasin D (Cyto D) an actin polymerization inhibitor. Next, to understand the mechanism, we assessed whether phagocytosis of fibers by mesothelial cells is affected by actin polymerization inhibition. Transmission electron microscopy showed the inhibition of fiber uptake by mesothelial cells in the presence of Cyto D. Furthermore, localization of components of the inflammasome, apoptotic speck-like protein containing a CARD domain (ASC) and NLRP3, to the perinuclear space in mitochondria or endoplasmic reticulum in response to fiber exposure was also interrupted in the presence of Cyto D. Taken together, our studies suggest that actin polymerization plays important roles in inflammasome activation by fibers via regulation of phagocytosis and/or spatial localization of inflammasome components.


Subject(s)
Actins/metabolism , Asbestos/adverse effects , Epithelial Cells/drug effects , Inflammasomes/drug effects , Actins/antagonists & inhibitors , Asbestos/antagonists & inhibitors , Cell Survival/drug effects , Cells, Cultured , Cytochalasin D/pharmacology , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Humans , Inflammasomes/metabolism , Polymerization/drug effects , Structure-Activity Relationship
12.
Cancer Prev Res (Phila) ; 9(5): 406-414, 2016 05.
Article in English | MEDLINE | ID: mdl-26935421

ABSTRACT

Exposure to asbestos is causally associated with the development of malignant mesothelioma, a cancer of cells lining the internal body cavities. Malignant mesothelioma is an aggressive cancer resistant to all current therapies. Once inhaled or ingested, asbestos causes inflammation in and around tissues that come in contact with these carcinogenic fibers. Recent studies suggest that inflammation is a major contributing factor in the development of many types of cancer, including malignant mesothelioma. The NALP3/NLRP3 inflammasome, including the component ASC, is thought to be an important mediator of inflammation in cells that sense extracellular insults, such as asbestos, and activate a signaling cascade resulting in release of mature IL1ß and recruitment of inflammatory cells. To determine if inflammasome-mediated inflammation contributes to asbestos-induced malignant mesothelioma, we chronically exposed Asc-deficient mice and wild-type littermates to asbestos and evaluated differences in tumor incidence and latency. The Asc-deficient mice showed significantly delayed tumor onset and reduced malignant mesothelioma incidence compared with wild-type animals. We also tested whether inflammation-related release of IL1ß contributes to tumor development in an accelerated mouse model of asbestos-induced malignant mesothelioma. Nf2(+/-);Cdkn2a(+/-) mice exposed to asbestos in the presence of anakinra, an IL1 receptor (IL1R) antagonist, showed a marked delay in the median time of malignant mesothelioma onset compared with similarly exposed mice given vehicle control (33.1 weeks vs. 22.6 weeks, respectively). Collectively, these studies provide evidence for a link between inflammation-related IL1ß/IL1R signaling and the development of asbestos-induced malignant mesothelioma. Furthermore, these findings provide rationale for chemoprevention strategies targeting IL1ß/IL1R signaling in high-risk, asbestos-exposed populations. Cancer Prev Res; 9(5); 406-14. ©2016 AACR.


Subject(s)
Inflammation/metabolism , Inflammation/pathology , Interleukin-1beta/metabolism , Lung Neoplasms/pathology , Mesothelioma/pathology , Receptors, Interleukin-1/metabolism , Animals , Apoptosis Regulatory Proteins/deficiency , Asbestos/adverse effects , CARD Signaling Adaptor Proteins , Disease Models, Animal , Humans , Immunoblotting , Immunohistochemistry , Lung Neoplasms/metabolism , Mesothelioma/metabolism , Mesothelioma, Malignant , Mice , Mice, Knockout , Signal Transduction/physiology
13.
PLoS One ; 10(12): e0145404, 2015.
Article in English | MEDLINE | ID: mdl-26689911

ABSTRACT

Malignant mesothelioma (MM) is a fatal disease in dire need of therapy. The role of inflammasomes in cancer is not very well studied, however, literature supports both pro-and anti-tumorigenic effects of inflammasomes on cancer depending upon the type of cancer. Asbestos is a causative agent for MM and we have shown before that it causes inflammasome priming and activation in mesothelial cells. MM tumor cells/tissues showed decreased levels of inflammasome components like NLRP3 and caspase-1 as compared to human mesothelial cells or normal tissue counterpart of tumor. Based on our preliminary findings we hypothesized that treatment of MMs with chemotherapeutic drugs may elevate the levels of NLRP3 and caspase-1 resulting in increased cell death by pyroptosis while increasing the levels of IL-1ß and other pro-inflammatory molecules. Therefore, a combined strategy of chemotherapeutic drug and IL-1R antagonist may play a beneficial role in MM therapy. To test our hypothesis we used two human MM tumor cell lines (Hmeso, H2373) and two chemotherapeutic drugs (doxorubicin, cisplatin). Through a series of experiments we showed that both chemotherapeutic drugs caused increases in NLRP3 levels, caspase-1 activation, pyroptosis and pro-inflammatory molecules released from MM cells. In vivo studies using SCID mice and Hmeso cells showed that tumors were smaller in combined treatment group of cisplatin and IL-1R antagonist (Anakinra) as compared to cisplatin alone or untreated control groups. Taken together our study suggests that chemotherapeutic drugs in combination with IL-1R antagonist may have a beneficial role in MM treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carrier Proteins/metabolism , Inflammasomes/drug effects , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Animals , Carrier Proteins/genetics , Caspase 1/metabolism , Cell Line, Tumor/drug effects , Cisplatin/administration & dosage , Cisplatin/pharmacology , Doxorubicin/pharmacology , Feedback, Physiological/drug effects , Humans , Inflammasomes/metabolism , Interleukin 1 Receptor Antagonist Protein/administration & dosage , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mesothelioma/metabolism , Mesothelioma/pathology , Mesothelioma, Malignant , Mice, SCID , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/metabolism , Xenograft Model Antitumor Assays
14.
Environ Health Perspect ; 123(8): A194-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26230287

ABSTRACT

Asbestos-related diseases continue to result in approximately 120,000 deaths every year in the United States and worldwide. Although extensive research has been conducted on health effects of occupational exposures to asbestos, many issues related to environmental asbestos exposures remain unresolved. For example, environmental asbestos exposures associated with a former mine in Libby, Montana, have resulted in high rates of nonoccupational asbestos-related disease. Additionally, other areas with naturally occurring asbestos deposits near communities in the United States and overseas are undergoing investigations to assess exposures and potential health risks. Some of the latest public health, epidemiological, and basic research findings were presented at a workshop on asbestos at the 2014 annual meeting of the Society of Toxicology in Phoenix, Arizona. The following focus areas were discussed: a) mechanisms resulting in fibrosis and/or tumor development; b) relative toxicity of different forms of asbestos and other hazardous elongated mineral particles (EMPs); c) proper dose metrics (e.g., mass, fiber number, or surface area of fibers) when interpreting asbestos toxicity; d) asbestos exposure to susceptible populations; and e) using toxicological findings for risk assessment and remediation efforts. The workshop also featured asbestos research supported by the National Institute of Environmental Health Sciences, the Agency for Toxic Substances and Disease Registry, and the U.S. Environmental Protection Agency. Better protection of individuals from asbestos-related health effects will require stimulation of new multidisciplinary research to further our understanding of what constitutes hazardous exposures and risk factors associated with toxicity of asbestos and other hazardous EMPs (e.g., nanomaterials).


Subject(s)
Asbestos/toxicity , Asbestosis/etiology , Environmental Exposure , Environmental Pollutants/toxicity , Environmental Restoration and Remediation , Humans , Risk Assessment
15.
PLoS One ; 10(5): e0127310, 2015.
Article in English | MEDLINE | ID: mdl-26011724

ABSTRACT

Dysregulation of signaling pathways and energy metabolism in cancer cells enhances production of mitochondrial hydrogen peroxide that supports tumorigenesis through multiple mechanisms. To counteract the adverse effects of mitochondrial peroxide many solid tumor types up-regulate the mitochondrial thioredoxin reductase 2--thioredoxin 2 (TRX2)--peroxiredoxin 3 (PRX3) antioxidant network. Using malignant mesothelioma cells as a model, we show that thiostrepton (TS) irreversibly disables PRX3 via covalent crosslinking of peroxidatic and resolving cysteine residues in homodimers, and that targeting the oxidoreductase TRX2 with the triphenylmethane gentian violet (GV) potentiates adduction by increasing levels of disulfide-bonded PRX3 dimers. Due to the fact that activity of the PRX3 catalytic cycle dictates the rate of adduction by TS, immortalized and primary human mesothelial cells are significantly less sensitive to both compounds. Moreover, stable knockdown of PRX3 reduces mesothelioma cell proliferation and sensitivity to TS. Expression of catalase in shPRX3 mesothelioma cells restores defects in cell proliferation but not sensitivity to TS. In a SCID mouse xenograft model of human mesothelioma, administration of TS and GV together reduced tumor burden more effectively than either agent alone. Because increased production of mitochondrial hydrogen peroxide is a common phenotype of malignant cells, and TS and GV are well tolerated in mammals, we propose that targeting PRX3 is a feasible redox-dependent strategy for managing mesothelioma and other intractable human malignancies.


Subject(s)
Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mesothelioma/drug therapy , Mesothelioma/metabolism , Mitochondria/metabolism , Peroxides/metabolism , Peroxiredoxin III/metabolism , Thiostrepton/pharmacology , Animals , Catalase/metabolism , Cell Proliferation/drug effects , Epithelium/drug effects , Epithelium/metabolism , Humans , Male , Mesothelioma, Malignant , Mice , Mice, SCID , Mitochondria/drug effects , Oxidation-Reduction/drug effects , Rats , Signal Transduction/drug effects , Signal Transduction/physiology , Thioredoxins/metabolism
16.
J Cell Biochem ; 116(8): 1540-52, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25757056

ABSTRACT

Malignant mesothelioma (MM) is an aggressive cancer of mesothelial cells of pleural and peritoneal cavities. In 85% of cases both pleural and peritoneal MM is caused by asbestos exposure. Although both are asbestos-induced cancers, the incidence of pleural MM is significantly higher (85%) than peritoneal MM (15%). It has been proposed that carcinogenesis is a result of asbestos-induced inflammation but it is not clear what contributes to the differences observed between incidences of these two cancers. We hypothesize that the observed differences in incidences of pleural and peritoneal MM are the result of differences in the direct response of these cell types to asbestos rather than to differences mediated by the in vivo microenvironment. To test this hypothesis we characterized cellular responses to asbestos in a controlled environment. We found significantly greater changes in genome-wide expression in response to asbestos exposure in pleural mesothelial cells as compared to peritoneal mesothelial cells. In particular, a greater response in many common genes (IL-8, ATF3, CXCL2, CXCL3, IL-6, GOS2) was seen in pleural mesothelial cells as compared to peritoneal mesothelial cells. Unique genes expressed in pleural mesothelial cells were mainly pro-inflammatory (G-CSF, IL-1ß, IL-1α, GREM1) and have previously been shown to be involved in development of MM. Our results are consistent with the hypothesis that differences in incidences of pleural and peritoneal MM upon exposure to asbestos are the result of differences in mesothelial cell physiology that lead to differences in the inflammatory response, which leads to cancer.


Subject(s)
Asbestos, Crocidolite/toxicity , Gene Expression Regulation, Neoplastic/drug effects , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mesothelioma/genetics , Mesothelioma/pathology , Peritoneal Neoplasms/genetics , Pleural Neoplasms/genetics , Adult , Aged , Cell Line , Cell Survival/drug effects , Female , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Inflammation/genetics , Lung Neoplasms/chemically induced , Male , Mesothelioma/chemically induced , Mesothelioma, Malignant , Middle Aged , Peritoneal Neoplasms/chemically induced , Peritoneal Neoplasms/pathology , Pleural Neoplasms/chemically induced , Pleural Neoplasms/pathology , Sequence Analysis, RNA
17.
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.

18.
Medicines (Basel) ; 2(4): 310-327, 2015 Dec.
Article in English | MEDLINE | ID: mdl-27088079

ABSTRACT

Exosomes are membrane-bound, intercellular communication shuttles that are defined by their endocytic origin and size range of 30-140 nm. Secreted by nearly all mammalian cell types and present in myriad bodily fluids, exosomes confer messages between cells, proximal and distal, by transporting biofunctional cargo in the form of proteins, nucleic acids, and lipids. They play a vital role in cellular signaling in both normal physiology and disease states, particularly cancer. Exosomes are powerful progenitors in altering target cell phenotypes, particularly in tumorigenesis and cancer progression, with the ability to alter tumor microenvironments and to assist in establishing the pre-metastatic niche. Many aspects of exosomes present them as novel means to identify cancer biomarkers for early detection and therapeutic targets, and using intrinsic and engineered characteristics of exosomes as therapeutic devices to ameliorate the progression of the disease. This review outlines some of the recent and major findings with regard to exosomes in cancer, and their utilization as therapeutic tools.

19.
Hum Mol Genet ; 24(5): 1374-89, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25351596

ABSTRACT

Occupational and environmental exposures to airborne asbestos and silica are associated with the development of lung fibrosis in the forms of asbestosis and silicosis, respectively. However, both diseases display distinct pathologic presentations, likely associated with differences in gene expression induced by different mineral structures, composition and bio-persistent properties. We hypothesized that effects of mineral exposure in the airway epithelium may dictate deviating molecular events that may explain the different pathologies of asbestosis versus silicosis. Using robust gene expression-profiling in conjunction with in-depth pathway analysis, we assessed early (24 h) alterations in gene expression associated with crocidolite asbestos or cristobalite silica exposures in primary human bronchial epithelial cells (NHBEs). Observations were confirmed in an immortalized line (BEAS-2B) by QRT-PCR and protein assays. Utilization of overall gene expression, unsupervised hierarchical cluster analysis and integrated pathway analysis revealed gene alterations that were common to both minerals or unique to either mineral. Our findings reveal that both minerals had potent effects on genes governing cell adhesion/migration, inflammation, and cellular stress, key features of fibrosis. Asbestos exposure was most specifically associated with aberrant cell proliferation and carcinogenesis, whereas silica exposure was highly associated with additional inflammatory responses, as well as pattern recognition, and fibrogenesis. These findings illustrate the use of gene-profiling as a means to determine early molecular events that may dictate pathological processes induced by exogenous cellular insults. In addition, it is a useful approach for predicting the pathogenicity of potentially harmful materials.


Subject(s)
Asbestos, Crocidolite/toxicity , Epithelial Cells/drug effects , Gene Expression Profiling , Lung/drug effects , Silicon Dioxide/toxicity , Carcinogenesis/chemically induced , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cluster Analysis , Dose-Response Relationship, Drug , Epithelial Cells/pathology , Humans , Inflammation/chemically induced , Inflammation/pathology , Lung/cytology , Microarray Analysis , Signal Transduction
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL