Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Ann Am Thorac Soc ; 20(8): 1077-1087, 2023 08.
Article in English | MEDLINE | ID: mdl-37526479

ABSTRACT

Rationale: To identify barriers and opportunities for Ph.D., basic and translational scientists to be fully integrated into clinical units. Objectives: In 2022, an ad hoc committee of the American Thoracic Society developed a project proposal and workshop to identify opportunities and barriers for scientists who do not practice medicine to develop successful careers and achieve tenure-track faculty positions in clinical departments and divisions within academic medical centers (AMCs) in the United States. Methods: This document focuses on results from a survey of adult and pediatric pulmonary, critical care, and sleep medicine division chiefs as well as a survey of workshop participants, including faculty in departmental and school leadership roles in both basic science and clinical units within U.S. AMCs. Results: We conclude that full integration of non-clinically practicing basic and translational scientists into the clinical units, in addition to their traditional placements in basic science units, best serves the tripartite mission of AMCs to provide care, perform research, and educate the next generation. Evidence suggests clinical units do employ Ph.D. scientists in large numbers, but these faculty are often hired into non-tenure track positions, which do not provide the salary support, start-up funds, research independence, or space often associated with hiring in basic science units within the same institution. These barriers to success of Ph.D. faculty in clinical units are largely financial. Conclusions: Our recommendation is for AMCs to consider and explore some of our proposed strategies to accomplish the goal of integrating basic and translational scientists into clinical units in a meaningful way.


Subject(s)
Academic Medical Centers , Physicians , Adult , United States , Humans , Child , Personnel Selection , Leadership , Faculty, Medical
2.
Cell Chem Biol ; 28(12): 1716-1727.e6, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34289376

ABSTRACT

GAS41 is an emerging oncogene overexpressed and implicated in multiple cancers, including non-small cell lung cancer (NSCLC). GAS41 is a dimeric protein that contains the YEATS domain, which is involved in the recognition of lysine-acylated histones. Here, we report the development of GAS41 YEATS inhibitors by employing a fragment-based screening approach. These inhibitors bind to GAS41 YEATS domain in a channel constituting a recognition site for acylated lysine on histone proteins. To enhance inhibitory activity, we developed a dimeric analog with nanomolar activity that blocks interactions of GAS41 with acetylated histone H3. Our lead compound engages GAS41 in cells, blocks proliferation of NSCLC cells, and modulates expression of GAS41-dependent genes, validating on-target mechanism of action. This study demonstrates that disruption of GAS41 protein-protein interactions may represent an attractive approach to target lung cancer cells. This work exemplifies the use of bivalent inhibitors as a general strategy to block challenging protein-protein interactions.


Subject(s)
Amides/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Thiophenes/pharmacology , Transcription Factors/antagonists & inhibitors , Amides/chemistry , Antineoplastic Agents/chemistry , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Cells, Cultured , Drug Screening Assays, Antitumor , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Molecular Structure , Protein Interaction Domains and Motifs/drug effects , Thiophenes/chemistry , Transcription Factors/metabolism
3.
Adv Sci (Weinh) ; 8(6): 2003747, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33747745

ABSTRACT

As the recognition between natural killer (NK) cells and cancer cells does not require antigen presentation, NK cells are being actively studied for use in adoptive cell therapies in the rapidly evolving armamentarium of cancer immunotherapy. In addition to utilizing NK cells, recent studies have shown that exosomes derived from NK cells also exhibit antitumor properties. Furthermore, these NK cell-derived exosomes exhibit higher stability, greater modification potentials and less immunogenicity compared to NK cells. Therefore, technologies that allow highly sensitive and specific isolation of NK cells and NK cell-derived exosomes can enable personalized NK-mediated cancer therapeutics in the future. Here, a novel microfluidic system to collect patient-specific NK cells and on-chip biogenesis of NK-exosomes is proposed. In a small cohort of non-small cell lung cancer (NSCLC) patients, both NK cells and circulating tumor cells (CTCs) were isolated, and it is found NSCLC patients have high numbers of NK and NK-exosomes compared with healthy donors, and these concentrations show a trend of positive and negative correlations with bloodborne CTC numbers, respectively. It is further demonstrated that the NK-exosomes harvested from NK-graphene oxide chip exhibit cytotoxic effect on CTCs. This versatile system is expected to be used for patient-specific NK-based immunotherapies along with CTCs for potential prognostic/diagnostic applications.

4.
JCI Insight ; 6(5)2021 03 08.
Article in English | MEDLINE | ID: mdl-33497367

ABSTRACT

Limitations of checkpoint inhibitor cancer immunotherapy include induction of autoimmune syndromes and resistance of many cancers. Since CD318, a novel CD6 ligand, is associated with the aggressiveness and metastatic potential of human cancers, we tested the effect of an anti-CD6 monoclonal antibody, UMCD6, on killing of cancer cells by human lymphocytes. UMCD6 augmented killing of breast, lung, and prostate cancer cells through direct effects on both CD8+ T cells and NK cells, increasing cancer cell death and lowering cancer cell survival in vitro more robustly than monoclonal antibody checkpoint inhibitors that interrupt the programmed cell death 1 (PD-1)/PD-1 ligand 1 (PD-L1) axis. UMCD6 also augmented in vivo killing by human peripheral blood lymphocytes of a human breast cancer line xenotransplanted into immunodeficient mice. Mechanistically, UMCD6 upregulated the expression of the activating receptor NKG2D and downregulated expression of the inhibitory receptor NKG2A on both NK cells and CD8+ T cells, with concurrent increases in perforin and granzyme B production. The combined capability of an anti-CD6 monoclonal antibody to control autoimmunity through effects on CD4+ lymphocyte differentiation while enhancing killing of cancer cells through distinct effects on CD8+ and NK cells opens a potential new approach to cancer immunotherapy that would suppress rather than instigate autoimmunity.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , CD8-Positive T-Lymphocytes/immunology , Immunotherapy/methods , Killer Cells, Natural/immunology , Neoplasms/therapy , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Line, Tumor , Humans , Killer Cells, Natural/cytology , Mice , Mice, SCID
6.
JCI Insight ; 4(20)2019 10 17.
Article in English | MEDLINE | ID: mdl-31619584

ABSTRACT

Lung cancer remains the leading cause of cancer-related death in the United States. Although the alveolar macrophage (AM) comprises the major resident immune cell in the lung, few studies have investigated its role in lung cancer development. We recently discovered a potentially novel mechanism wherein AMs regulate STAT-induced inflammatory responses in neighboring epithelial cells (ECs) via secretion and delivery of suppressors of cytokine signaling 3 (SOCS3) within extracellular vesicles (EVs). Here, we explored the impact of SOCS3 transfer on EC tumorigenesis and the integrity of AM SOCS3 secretion during development of lung cancer. AM-derived EVs containing SOCS3 inhibited STAT3 activation as well as proliferation and survival of lung adenocarcinoma cells. Levels of secreted SOCS3 were diminished in lungs of patients with non-small cell lung cancer and in a mouse model of lung cancer, and the impaired ability of murine AMs to secrete SOCS3 within EVs preceded the development of lung tumors. Loss of this homeostatic brake on tumorigenesis prompted our effort to "rescue" it. Provision of recombinant SOCS3 loaded within synthetic liposomes inhibited proliferation and survival of lung adenocarcinoma cells in vitro as well as malignant transformation of normal ECs. Intratumoral injection of SOCS3 liposomes attenuated tumor growth in a lung cancer xenograft model. This work identifies AM-derived vesicular SOCS3 as an endogenous antitumor mechanism that is disrupted within the tumor microenvironment and whose rescue by synthetic liposomes can be leveraged as a potential therapeutic strategy for lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Lung Neoplasms/immunology , Macrophages, Alveolar/immunology , Suppressor of Cytokine Signaling 3 Protein/metabolism , A549 Cells , Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/metabolism , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Carcinogenesis/drug effects , Carcinogenesis/immunology , Carcinogenesis/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Female , Humans , Injections, Intralesional , Liposomes , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Macrophages, Alveolar/cytology , Macrophages, Alveolar/metabolism , Mice , Primary Cell Culture , Rats , Recombinant Proteins/administration & dosage , Respiratory Mucosa/cytology , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein/administration & dosage , Suppressor of Cytokine Signaling 3 Protein/genetics , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
7.
Proc Natl Acad Sci U S A ; 116(1): 199-204, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30559191

ABSTRACT

Cysteinyl leukotrienes (cys-LTs) are proinflammatory mediators that enhance vascular permeability through distinct receptors (CysLTRs). We found that CysLT2R regulates angiogenesis in isolated mouse endothelial cells (ECs) and in Matrigel implants in WT mice and enhances EC contraction and permeability via the Rho-dependent myosin light chain 2 and vascular endothelial (VE)-cadherin axis. Since solid tumors utilize aberrant angiogenesis for their growth and metastasis and their vessels exhibit vascular hyperpermeability, we hypothesized that CysLT2R, via its actions on the endothelium, might regulate tumor growth. Both tumor growth and metastases of adoptively transferred syngeneic Lewis lung carcinoma (LLC) cells are significantly reduced in CysLT2R-null mice (Cysltr2-/-) compared with WT and CysLT1R-null mice (Cysltr1-/-). In WT recipients of LLC cells, CysLT2R expression is significantly increased in the tumor vasculature, compared with CysLT1R. Further, the tumor vasculature in Cysltr2-/- recipients exhibited significantly improved integrity, as revealed by increased pericyte coverage and decreased leakage of i.v.-administered Texas Red-conjugated dextran. Administration of a selective CysLT2R antagonist significantly reduced LLC tumor volume, vessel density, dextran leakage, and metastases in WT mice, highlighting CysLT2R as a VEGF-independent regulator of the vasculature promoting risk of metastasis. Thus, both genetic and pharmacological findings establish CysLT2R as a gateway for angiogenesis and EC dysregulation in vitro and ex vivo and in an in vivo model with a mouse tumor. Our data suggest CysLT2R as a possible target for intervention.


Subject(s)
Endothelial Cells/drug effects , Neovascularization, Pathologic/chemically induced , Receptors, Leukotriene/metabolism , Animals , Capillary Permeability/drug effects , Cyclohexanecarboxylic Acids/pharmacology , Gene Knockout Techniques , Leukotriene Antagonists/pharmacology , Lung Neoplasms/drug therapy , Male , Mice , Mice, Inbred C57BL , Neoplasm Metastasis/drug therapy , Neoplasm Transplantation , Neoplasms, Experimental , Neovascularization, Pathologic/drug therapy , Phthalic Acids/pharmacology , Receptors, Leukotriene/drug effects
9.
J Clin Invest ; 128(4): 1384-1396, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29324443

ABSTRACT

During epithelial-mesenchymal transition (EMT) epithelial cancer cells transdifferentiate into highly motile, invasive, mesenchymal-like cells, giving rise to disseminating tumor cells. Few of these disseminated cells successfully metastasize. Immune cells and inflammation in the tumor microenvironment were shown to drive EMT, but few studies investigated the consequences of EMT for tumor immunosurveillance. In addition to initiating metastasis, we demonstrate that EMT confers increased susceptibility to natural killer (NK) cells and contributes, in part, to the inefficiency of the metastatic process. Depletion of NK cells allowed spontaneous metastasis without affecting primary tumor growth. EMT-induced modulation of E-cadherin and cell adhesion molecule 1 (CADM1) mediated increased susceptibility to NK cytotoxicity. Higher CADM1 expression correlates with improved patient survival in 2 lung and 1 breast adenocarcinoma patient cohorts and decreased metastasis. Our observations reveal a novel NK-mediated, metastasis-specific immunosurveillance in lung cancer and present a window of opportunity for preventing metastasis by boosting NK cell activity.


Subject(s)
Carcinoma, Lewis Lung/immunology , Epithelial-Mesenchymal Transition/immunology , Immunity, Cellular , Immunologic Surveillance , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Tumor Microenvironment/immunology , A549 Cells , Animals , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Cell Adhesion Molecule-1/genetics , Cell Adhesion Molecule-1/immunology , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Killer Cells, Natural/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology
10.
Biochem Biophys Res Commun ; 489(2): 130-134, 2017 07 22.
Article in English | MEDLINE | ID: mdl-28536076

ABSTRACT

Arginine, a cationic amino acid is known to stabilize proteins under harsh conditions. It is widely used to stabilize protein aggregation, and to correct protein folding during protein production. Hence it would be a good therapeutic candidate for treating protein aggregation related diseases. Recent reports suggest, that the aggregation of tumor suppressor protein p53 is one of the leading causes of tumor progression. When mutated, p53 protein aggregates, loses its function leading to unwanted cell growth and ultimately results in tumor. Here in this study we focus on the inhibitory effects of polyarginine and its analogues polyornithine, canavanine, and citrulline on the inhibition of p53 mutant peptide aggregation, and p53 mutant cancer cell proliferation inhibition in vitro. Biochemical assays and cell toxicity studies were used to characterize the study. The results show that polyarginine, and polyornithine, in micromolar concentrations, significantly inhibits p53 conserved peptide aggregation, and the cell proliferation of p53 mutant cancer cells. Hence they could be promising candidates for treating p53 mutant/misfolded protein aggregation associated cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/genetics , Neoplasms/pathology , Peptides/pharmacology , Protein Aggregation, Pathological/drug therapy , Protein Aggregation, Pathological/prevention & control , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , High-Throughput Screening Assays , Humans , Mutation , Peptides/administration & dosage , Peptides/chemistry , Protein Aggregates/drug effects , Protein Aggregation, Pathological/pathology , Structure-Activity Relationship
11.
BMC Cancer ; 16: 614, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27501846

ABSTRACT

BACKGROUND: Circulating tumor cells (CTCs) have shown prognostic relevance in many cancer types. However, the majority of current CTC capture methods rely on positive selection techniques that require a priori knowledge about the surface protein expression of disseminated CTCs, which are known to be a dynamic population. METHODS: We developed a microfluidic CTC capture chip that incorporated a nanoroughened glass substrate for capturing CTCs from blood samples. Our CTC capture chip utilized the differential adhesion preference of cancer cells to nanoroughened etched glass surfaces as compared to normal blood cells and thus did not depend on the physical size or surface protein expression of CTCs. RESULTS: The microfluidic CTC capture chip was able to achieve a superior capture yield for both epithelial cell adhesion molecule positive (EpCAM+) and EpCAM- cancer cells in blood samples. Additionally, the microfluidic CTC chip captured CTCs undergoing transforming growth factor beta-induced epithelial-to-mesenchymal transition (TGF-ß-induced EMT) with dynamically down-regulated EpCAM expression. In a mouse model of human breast cancer using EpCAM positive and negative cell lines, the number of CTCs captured correlated positively with the size of the primary tumor and was independent of their EpCAM expression. Furthermore, in a syngeneic mouse model of lung cancer using cell lines with differential metastasis capability, CTCs were captured from all mice with detectable primary tumors independent of the cell lines' metastatic ability. CONCLUSIONS: The microfluidic CTC capture chip using a novel nanoroughened glass substrate is broadly applicable to capturing heterogeneous CTC populations of clinical interest independent of their surface marker expression and metastatic propensity. We were able to capture CTCs from a non-metastatic lung cancer model, demonstrating the potential of the chip to collect the entirety of CTC populations including subgroups of distinct biological and phenotypical properties. Further exploration of the biological potential of metastatic and presumably non-metastatic CTCs captured using the microfluidic chip will yield insights into their relevant differences and their effects on tumor progression and cancer outcomes.


Subject(s)
Cell Separation/methods , Epithelial Cell Adhesion Molecule/metabolism , Microfluidic Analytical Techniques/methods , Neoplasms/metabolism , Neoplastic Cells, Circulating/pathology , Transforming Growth Factor beta/pharmacology , A549 Cells , Animals , Cell Adhesion , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Female , Genetic Heterogeneity , Humans , MCF-7 Cells , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Neoplasms/pathology , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/metabolism
12.
J Immunol ; 197(3): 691-8, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27431984

ABSTRACT

Microenvironments that tumor cells encounter are different during the stages of cancer progression-primary tumor, metastasis, and at the metastatic site. This suggests potential differences in immune surveillance of primary tumor and metastasis. Epithelial-mesenchymal transition (EMT) is a key reversible process in which cancer cells transition into highly motile and invasive cells for dissemination. Only a tiny proportion successfully metastasize, supporting the notion of metastasis-specific immune surveillance. EMT involves extensive molecular reprogramming of cells conferring many clinically relevant features to cancer cells and affects tumor cell interactions within the tumor microenvironment. We review the impact of tumor immune infiltrates on tumor cell EMT and the consequences of EMT in shaping the immune microenvironment of tumors. The usefulness of EMT as a model to investigate metastasis-specific immune surveillance mechanisms are also explored. Finally, we discuss potential implications of EMT for tumor immunogenicity, as well as current immunotherapies and future strategies.


Subject(s)
Epithelial-Mesenchymal Transition/immunology , Neoplasms/pathology , Tumor Escape/immunology , Tumor Microenvironment/immunology , Animals , Disease Progression , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/immunology
13.
Am J Pathol ; 185(4): 969-86, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25681733

ABSTRACT

Myofibroblasts are crucial to the pathogenesis of tissue fibrosis. Their formation of stress fibers results in the release of myocardin-related transcription factor (MRTF), a transcriptional coactivator of serum response factor (SRF). MRTF-A (Mkl1)-deficient mice are protected from lung fibrosis. We hypothesized that the SRF/MRTF pathway inhibitor CCG-203971 would modulate myofibroblast function in vitro and limit lung fibrosis in vivo. Normal and idiopathic pulmonary fibrosis lung fibroblasts were treated with/without CCG-203971 (N-[4-chlorophenyl]-1-[3-(2-furanyl)benzoyl]-3-piperidine carboxamide) and/or Fas-activating antibody in the presence/absence of transforming growth factor (TGF)-ß1, and apoptosis was assessed. In vivo studies examined the effect of therapeutically administered CCG-203971 on lung fibrosis in two distinct murine models of fibrosis induced by bleomycin or targeted type II alveolar epithelial injury. In vitro, CCG-203971 prevented nuclear localization of MRTF-A; increased the apoptotic susceptibility of normal and idiopathic pulmonary fibrosis fibroblasts; blocked TGF-ß1-induced myofibroblast differentiation; and inhibited TGF-ß1-induced expression of fibronectin, X-linked inhibitor of apoptosis, and plasminogen activator inhibitor-1. TGF-ß1 did not protect fibroblasts or myofibroblasts from apoptosis in the presence of CCG-203971. In vivo, CCG-203971 significantly reduced lung collagen content in both murine models while decreasing alveolar plasminogen activator inhibitor-1 and promoting myofibroblast apoptosis. These data support a central role of the SRF/MRTF pathway in the pathobiology of lung fibrosis and suggest that its inhibition can help resolve lung fibrosis by promoting fibroblast apoptosis.


Subject(s)
Apoptosis , Lung/metabolism , Lung/pathology , Mesoderm/pathology , Serum Response Factor/metabolism , Signal Transduction , Trans-Activators/metabolism , Adult , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytoprotection/drug effects , Fibronectins/metabolism , Fibrosis , Humans , Inflammation/pathology , Mesoderm/drug effects , Mice, Inbred C57BL , Myofibroblasts/pathology , Nipecotic Acids/administration & dosage , Nipecotic Acids/pharmacology , Plasminogen Activator Inhibitor 1/metabolism , Protein Transport/drug effects , Signal Transduction/drug effects , Sus scrofa , Transforming Growth Factor beta1/pharmacology , X-Linked Inhibitor of Apoptosis Protein/metabolism , fas Receptor/metabolism
14.
Carcinogenesis ; 35(6): 1292-300, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24510113

ABSTRACT

In cancer cells, the process of epithelial-mesenchymal transition (EMT) confers migratory and invasive capacity, resistance to apoptosis, drug resistance, evasion of host immune surveillance and tumor stem cell traits. Cells undergoing EMT may represent tumor cells with metastatic potential. Characterizing the EMT secretome may identify biomarkers to monitor EMT in tumor progression and provide a prognostic signature to predict patient survival. Utilizing a transforming growth factor-ß-induced cell culture model of EMT, we quantitatively profiled differentially secreted proteins, by GeLC-tandem mass spectrometry. Integrating with the corresponding transcriptome, we derived an EMT-associated secretory phenotype (EASP) comprising of proteins that were differentially upregulated both at protein and mRNA levels. Four independent primary tumor-derived gene expression data sets of lung cancers were used for survival analysis by the random survival forests (RSF) method. Analysis of 97-gene EASP expression in human lung adenocarcinoma tumors revealed strong positive correlations with lymph node metastasis, advanced tumor stage and histological grade. RSF analysis built on a training set (n = 442), including age, sex and stage as variables, stratified three independent lung cancer data sets into low-, medium- and high-risk groups with significant differences in overall survival. We further refined EASP to a 20 gene signature (rEASP) based on variable importance scores from RSF analysis. Similar to EASP, rEASP predicted survival of both adenocarcinoma and squamous carcinoma patients. More importantly, it predicted survival in the early-stage cancers. These results demonstrate that integrative analysis of the critical biological process of EMT provides mechanism-based and clinically relevant biomarkers with significant prognostic value.


Subject(s)
Epithelial-Mesenchymal Transition , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Phenotype , Adult , Aged , Cell Line, Tumor , Cluster Analysis , Computational Biology , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression , Gene Expression Profiling , Humans , Lung Neoplasms/mortality , Lung Neoplasms/therapy , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis , Proteomics
15.
Oncoimmunology ; 1(3): 340-345, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22737610

ABSTRACT

Tumors arise and progress in immunocompetent hosts presumably by activating tolerance mechanisms critical for normal homeostasis. Host immune cells can mount anti-tumor responses by activation of Toll-like receptors (TLRs). However, emerging data suggests that molecules that negatively regulate TLRs are exploited by tumors to induce tolerance and mitigate the host immunosurveillance. Targeting these negative regulators can be a potential new immunotherapeutic strategy.

16.
J Thorac Oncol ; 6(11): 1784-92, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21964532

ABSTRACT

BACKGROUND: Acquisition of mesenchymal phenotype by epithelial cells by means of epithelial-mesenchymal transition (EMT) is considered as an early event in the multistep process of tumor metastasis. Therefore, inhibition of EMT might be a rational strategy to prevent metastasis. METHODS: Using the global gene expression profile from a cell culture model of transforming growth factor-ß (TGF-ß)-induced EMT, we identified potential EMT inhibitors. We used a publicly available database (www.broad.mit.edu/cmap) comprising gene expression profiles obtained from multiple different cell lines in response to various drugs to derive negative correlations to EMT gene expression profile using Connectivity Map, a pattern matching tool. RESULTS: Experimental validation of the identified compounds showed rapamycin as a novel inhibitor of TGF-ß signaling along with 17-AAG, a known modulator of TGF-ß pathway. Both of these compounds completely blocked EMT and the associated migratory and invasive phenotype. The other identified compound, LY294002, demonstrated a selective inhibition of mesenchymal markers, cell migration and invasion, without affecting the loss of E-cadherin expression or Smad phosphorylation. CONCLUSIONS: Our data reveal that rapamycin is a novel modulator of TGF-ß signaling, and along with 17-AAG and LY294002, could be used as therapeutic agent for inhibiting EMT. This study demonstrates the potential of a systems approach in identifying novel modulators of a complex biological process.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/pathology , Adenocarcinoma/pathology , Benzoquinones/pharmacology , Cell Movement/drug effects , Enzyme Inhibitors/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Lactams, Macrocyclic/pharmacology , Lung Neoplasms/pathology , Transforming Growth Factor beta/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Adenocarcinoma, Bronchiolo-Alveolar/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cadherins/metabolism , Chromones/pharmacology , Gene Expression Profiling , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Immunosuppressive Agents/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Morpholines/pharmacology , Oligonucleotide Array Sequence Analysis , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction , Sirolimus/pharmacology , Smad Proteins/metabolism , Transcription, Genetic , Tumor Cells, Cultured
17.
Am J Pathol ; 178(6): 2461-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21641374

ABSTRACT

Fibrotic obliteration of the small airways leading to progressive airflow obstruction, termed bronchiolitis obliterans syndrome (BOS), is the major cause of poor outcomes after lung transplantation. We recently demonstrated that a donor-derived population of multipotent mesenchymal stem cells (MSCs) can be isolated from the bronchoalveolar lavage (BAL) fluid of human lung transplant recipients. Herein, we study the organ specificity of these cells and investigate the role of local mesenchymal progenitors in fibrogenesis after lung transplantation. We demonstrate that human lung allograft-derived MSCs uniquely express embryonic lung mesenchyme-associated transcription factors with a 35,000-fold higher expression of forkhead/winged helix transcription factor forkhead box (FOXF1) noted in lung compared with bone marrow MSCs. Fibrotic differentiation of MSCs isolated from normal lung allografts was noted in the presence of profibrotic mediators associated with BOS, including transforming growth factor-ß and IL-13. MSCs isolated from patients with BOS demonstrated increased expression of α-SMA and collagen I when compared with non-BOS controls, consistent with a stable in vivo fibrotic phenotype. FOXF1 mRNA expression in the BAL cell pellet correlated with the number of MSCs in the BAL fluid, and myofibroblasts present in the fibrotic lesions expressed FOXF1 by in situ hybridization. These data suggest a key role for local tissue-specific, organ-resident, mesenchymal precursors in the fibrogenic processes in human adult lungs.


Subject(s)
Lung Transplantation , Lung/pathology , Mesenchymal Stem Cells/pathology , Actins/metabolism , Biomarkers/metabolism , Biopsy , Bone Marrow Cells/pathology , Bronchiolitis Obliterans/pathology , Bronchoalveolar Lavage Fluid , Cell Count , Cell Differentiation , Cell Separation , Collagen/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibrosis , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Humans , Lung/embryology , Lung/metabolism , Mesenchymal Stem Cells/metabolism , Myofibroblasts/pathology , Organ Specificity , Phenotype , Receptors, Interleukin-13/metabolism , Transplantation, Homologous
18.
Lung Cancer ; 72(2): 154-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21354647

ABSTRACT

Peroxisome proliferator-activated receptors (PPAR)-γ belongs to the nuclear hormone receptor superfamily of ligand-dependent transcription factors. It is a mediator of adipocyte differentiation, regulates lipid metabolism and macrophage function. The ligands of PPAR-γ have long been in the clinic for the treatment of type II diabetes and have a very low toxicity profile. Activation of PPAR-γ was shown to modulate various hallmarks of cancer through its pleiotropic affects on multiple different cell types in the tumor microenvironment. An overwhelming number of preclinical-studies demonstrate the efficacy of PPAR-γ ligands in the control of tumor progression through their affects on various cellular processes, including cell proliferation, apoptosis, angiogenesis, inflammation and metastasis. A variety of signaling pathways have been implicated as potential mechanisms of action. This review will focus on the molecular basis of these mechanisms; primarily PPAR-γ cross-regulation with other signaling pathways and its relevance to lung cancer therapy will be discussed.


Subject(s)
Antineoplastic Agents/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , PPAR gamma/agonists , Receptor Cross-Talk , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Drug Therapy/trends , Humans , Inflammation , Lung Neoplasms/pathology , Neoplasm Metastasis , Receptor Cross-Talk/drug effects , Signal Transduction/drug effects
19.
J Oncol ; 2011: 172678, 2011.
Article in English | MEDLINE | ID: mdl-22229030
20.
Mol Cancer Ther ; 9(12): 3221-32, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21159608

ABSTRACT

Epithelial-mesenchymal transition (EMT) was shown to confer tumor cells with abilities essential for metastasis, including migratory phenotype, invasiveness, resistance to apoptosis, evading immune surveillance, and tumor stem cell traits. Therefore, inhibition of EMT can be an important therapeutic strategy to inhibit tumor metastasis. Here, we show that activation of peroxisome proliferator-activated receptor γ (PPAR-γ) inhibits transforming growth factor ß (TGF-ß)-induced EMT in lung cancer cells and prevents metastasis by antagonizing Smad3 function. Activation of PPAR-γ by synthetic ligands (troglitazone and rosiglitazone) or by a constitutively active form of PPAR-γ prevents TGF-ß-induced loss of E-cadherin expression and inhibits the induction of mesenchymal markers (vimentin, N-cadherin, fibronectin) and matrix metalloproteases. Consistently, activation of PPAR-γ also inhibited EMT-induced migration and invasion of lung cancer cells. Furthermore, effects of PPAR-γ ligands were attenuated by siRNA-mediated knockdown of PPAR-γ, indicating that the ligand-induced responses are PPAR-γ dependent. Selective knockdown of Smad2 and Smad3 by siRNA showed that TGF-ß-induced EMT is Smad3 dependent in lung cancer cells. Activation of PPAR-γ inhibits TGF-ß-induced Smad transcriptional activity but had no effect on the phosphorylation or nuclear translocation of Smads. Consistently, PPAR-γ activation prevented TGF-ß-induced transcriptional repression of E-cadherin promoter and inhibited transcriptional activation of N-cadherin promoter. Finally, treatment of mice with troglitazone or knockdown of Smad3 in tumor cells significantly inhibited TGF-ß-induced experimental metastasis in SCID-Beige mice. Together, with the low toxicity profile of PPAR-γ ligands, our data show that these ligands may serve as potential therapeutic agents to inhibit metastasis.


Subject(s)
Epithelial-Mesenchymal Transition , PPAR gamma/metabolism , Smad3 Protein/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Epithelial-Mesenchymal Transition/drug effects , Gene Knockdown Techniques , Humans , Ligands , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Neoplasm Metastasis , Phenotype , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Protein Transport/drug effects , Smad2 Protein/metabolism , Smad3 Protein/genetics , Transcription, Genetic/drug effects , Transforming Growth Factor beta/pharmacology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...