Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Proteome Res ; 14(8): 3039-50, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26120931

ABSTRACT

Lung cancer is the leading cause of cancer mortality worldwide. Although many biomarkers have been identified for lung cancer, their low specificity and sensitivity present an urgent need for the identification of more candidate biomarkers. In this study, we conducted MRM-based targeted analysis to evaluate the potential utility of a list of candidate proteins for lung cancer diagnosis. A total of 1249 transitions of 420 peptides representing 102 candidate proteins from our previous study and the literature were first screened by MRM analysis in pooled plasma samples, resulting in 78 proteins remaining in the list. Relative quantification of these 78 proteins was further performed in 60 individual plasma samples from lung adenocarcinoma patients in stages I-III and matched healthy control subjects. Ultimately, nine proteins were found to be able to distinguish patients from controls. Further combinations of five, three, and two candidate marker proteins improved the sensitivity to discriminate patients from controls and resulted in a merged AUC value of nearly 1.00 in stages I-III patients versus controls. Our results highlighted several possible markers for lung adenocarcinoma, and the proposed protein panels require further validation in a larger cohort to evaluate their potential use in clinical applications or development of therapeutics.


Subject(s)
Adenocarcinoma/blood , Biomarkers, Tumor/blood , Lung Neoplasms/blood , Mass Spectrometry/methods , Peptides/blood , Proteomics/methods , Adenocarcinoma/diagnosis , Adenocarcinoma/metabolism , Aged , Aged, 80 and over , Amino Acid Sequence , Biomarkers, Tumor/metabolism , Blood Proteins/analysis , Blood Proteins/metabolism , Female , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/metabolism , Male , Middle Aged , Molecular Sequence Data , Neoplasm Staging , Peptides/metabolism , Reproducibility of Results , Sensitivity and Specificity
2.
Proteomics ; 15(15): 2560-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26033914

ABSTRACT

Intramyocardial transplantation of cardiomyocytes (CMs), endothelial cells (ECs), and smooth muscle cells (SMCs) derived from human induced pluripotent stem cells (hiPSCs) has beneficial effects on the post-infarction heart. However, the mechanisms underlying the functional improvements remain undefined. We employed large-scale label-free quantitative proteomics to identify proteins that were differentially regulated following cellular transplantation in a swine model of myocardial infarction (MI). We identified 22 proteins that were significantly up-regulated after trilineage cell transplantation compared to both MI and Sham groups. Among them, 12 proteins, including adenylyl cyclase-associated protein 1 and tropomodulin-1, are associated with positive regulation of muscular contraction whereas 11 proteins, such as desmoplakin and zyxin, are involved in embryonic and muscular development and regeneration. Moreover, we identified 21 proteins up-regulated and another 21 down-regulated in MI, but reversed after trilineage cell transplantation. Proteins up-regulated after MI but reversed by transplantation are related to fibrosis and apoptosis. Conversely, proteins down-regulated in MI but restored after cell therapy are regulators of protein nitrosylation. Our results show that the functionally beneficial effects of trilineage cell therapy are accompanied by differential regulation of protein expression in the recipient myocardium, which may contribute to the improved cardiac function.


Subject(s)
Induced Pluripotent Stem Cells/transplantation , Myocardial Infarction/therapy , Proteomics/methods , Stem Cell Transplantation/methods , Animals , Blotting, Western , Cell Line , Cells, Cultured , Chromatography, Liquid/methods , Computational Biology/methods , Humans , Infant, Newborn , Male , Mice , Myocardium/metabolism , Swine , Tandem Mass Spectrometry , Transplantation, Heterologous
3.
J Proteome Res ; 14(3): 1587-99, 2015 Mar 06.
Article in English | MEDLINE | ID: mdl-25589168

ABSTRACT

Tissue proteomics is increasingly recognized for its role in biomarker discovery and disease mechanism investigation. However, protein solubility remains a significant challenge in mass spectrometry (MS)-based tissue proteomics. Conventional surfactants such as sodium dodecyl sulfate (SDS), the preferred surfactant for protein solubilization, are not compatible with MS. Herein, we have screened a library of surfactant-like compounds and discovered an MS-compatible degradable surfactant (MaSDeS) for tissue proteomics that solubilizes all categories of proteins with performance comparable to SDS. The use of MaSDeS in the tissue extraction significantly improves the total number of protein identifications from commonly used tissues, including tissue from the heart, liver, and lung. Notably, MaSDeS significantly enriches membrane proteins, which are often under-represented in proteomics studies. The acid degradable nature of MaSDeS makes it amenable for high-throughput MS-based proteomics. In addition, the thermostability of MaSDeS allows for its use in experiments requiring high temperature to facilitate protein extraction and solubilization. Furthermore, we have shown that MaSDeS outperforms the other MS-compatible surfactants in terms of overall protein solubility and the total number of identified proteins in tissue proteomics. Thus, the use of MaSDeS will greatly advance tissue proteomics and realize its potential in basic biomedical and clinical research. MaSDeS could be utilized in a variety of proteomics studies as well as general biochemical and biological experiments that employ surfactants for protein solubilization.


Subject(s)
Mass Spectrometry/methods , Proteomics , Surface-Active Agents/chemistry , Animals , Swine
4.
Tumour Biol ; 36(5): 3957-67, 2015 May.
Article in English | MEDLINE | ID: mdl-25578494

ABSTRACT

Tissue inhibitor metalloproteinase-1 (TIMP-1) is clinically associated with a poor prognosis for various cancers, but the roles of TIMP-1 in lung cancer metastasis are controversial. Our previous secretomic study revealed that TIMP-1 is highly abundant in high invasiveness cells of lung adenocarcinoma. In the current study, TIMP-1 abundances in primary lung adenocarcinoma tissues, as revealed by immunohistochemistry, are significantly higher in patients with lymph invasion and distant metastasis than in those without. Receiver operating characteristic curve analyses suggest 73.7 and 86.2 % accuracy to separate patients with lymph node and distant metastasis and those without, respectively. Moreover, we demonstrate that the expression level of TIMP-1 positively associates with cell mobility, invasiveness, and metastatic colonization. Most notably, the novel mechanism in which TIMP-1 facilitates metastatic colonization through the mediation of pericellular polyFN1 assembly was revealed. In summary, this study presents novel functions of TIMP-1 in promoting cancer metastasis and suggests TIMP-1 is a potential tissue biomarker for lymph invasion and distant metastasis of lung adenocarcinoma.


Subject(s)
Adenocarcinoma/genetics , Cell Movement/genetics , Lung Neoplasms/genetics , Neoplasm Invasiveness/genetics , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Adult , Aged , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis , Prognosis , Tissue Inhibitor of Metalloproteinase-1/genetics
5.
J Thorac Oncol ; 10(2): 286-301, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25299231

ABSTRACT

BACKGROUND: Differential expression and secretion of alpha-actinin 4 (ACTN4) in the lung cancer cell lines CL1-0 and CL1-5 have been reported in previous proteomic studies. The aim of this study is to investigate the functional properties of the ACTN4 protein in non-small-cell lung cancer (NSCLC) cells and evaluate its clinical importance. METHODS: We used RNA interference to knock down and overexpress ACTN4 protein to evaluate the effects of this intervention on cancer cell invasion and migration, as well as on microscopic cellular morphology. Furthermore, we examined by immunohistochemistry the expression of ACTN4 protein in tissue samples at different stages of lung cancer and compared the protein levels of ACTN4 in blood plasma samples from patients with histologically confirmed lung cancer and healthy controls. RESULTS: CL1-5 cell motility was significantly suppressed by the knockdown of ACTN4 protein. The morphology of CL1-5 cells changed from a predominantly mesenchymal-like shape into a globular shape in response to ACTN4 protein knockdown. A quantitative immunohistochemical assessment of lung cancer tissues revealed that ACTN4 protein level was considerably higher in cancerous tissues than in the adjacent normal ones, and the area under the receiver operating characteristic curve was 0.736 (p < 0.001). According to an enzyme-linked immunosorbent assay, the plasma levels of ACTN4 protein were significantly different between cancer patients and healthy controls, and the areas under the receiver operating characteristic curves were 0.828 and 0.909, respectively, for two independent cohorts (p < 0.001). CONCLUSIONS: We demonstrate that the knockdown of ACTN4 protein inhibited cell invasion and migration. These results suggest that ACTN4 is associated with lung cancer cell motility. Thus, the level of ACTN4 in cancerous tissue and plasma is related to the presence of lung cancer.


Subject(s)
Actinin/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Movement/physiology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Actinin/biosynthesis , Actinin/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Case-Control Studies , Cell Growth Processes/physiology , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Lung Neoplasms/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Transfection
6.
Cell Stem Cell ; 15(6): 750-61, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25479750

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) hold promise for myocardial repair following injury, but preclinical studies in large animal models are required to determine optimal cell preparation and delivery strategies to maximize functional benefits and to evaluate safety. Here, we utilized a porcine model of acute myocardial infarction (MI) to investigate the functional impact of intramyocardial transplantation of hiPSC-derived cardiomyocytes, endothelial cells, and smooth muscle cells, in combination with a 3D fibrin patch loaded with insulin growth factor (IGF)-encapsulated microspheres. hiPSC-derived cardiomyocytes integrated into host myocardium and generated organized sarcomeric structures, and endothelial and smooth muscle cells contributed to host vasculature. Trilineage cell transplantation significantly improved left ventricular function, myocardial metabolism, and arteriole density, while reducing infarct size, ventricular wall stress, and apoptosis without inducing ventricular arrhythmias. These findings in a large animal MI model highlight the potential of utilizing hiPSC-derived cells for cardiac repair.


Subject(s)
Endothelial Cells/transplantation , Heart Ventricles/metabolism , Induced Pluripotent Stem Cells/physiology , Myocardial Infarction/therapy , Myocardium/metabolism , Myocytes, Cardiac/transplantation , Myocytes, Smooth Muscle/transplantation , Stem Cell Transplantation , Acute Disease , Animals , Apoptosis , Cell Differentiation , Cell Lineage , Cells, Cultured , Disease Models, Animal , Endothelial Cells/physiology , Fibrin/administration & dosage , Heart Ventricles/pathology , Humans , Insulin-Like Growth Factor I/administration & dosage , Microspheres , Myocardial Infarction/pathology , Myocytes, Cardiac/physiology , Myocytes, Smooth Muscle/physiology , Recovery of Function , Swine
7.
Nat Commun ; 5: 4804, 2014 Sep 03.
Article in English | MEDLINE | ID: mdl-25183545

ABSTRACT

Rab small GTPases are master regulators of membrane trafficking and guide vesicle targeting. Recent publications show that Rab-controlled trafficking pathways are altered during tumorigenesis. However, whether any of the Rabs plays a metastasis suppressor role is least explored. Here we address the metastasis suppressive function of human Rab37 (hRAB37) using secretomics, cell, animal and clinical analyses. We show that tissue inhibitor of metalloproteinase 1 (TIMP1), a secreted glycoprotein that inhibits extracellular matrix turnover, is a novel cargo of hRAB37. hRAB37 regulates the exocytosis of TIMP1 in a nucleotide-dependent manner to inactivate matrix metalloproteinase 9 (MMP9) migration axis in vitro and in vivo. Dysfunction of hRAB37 or TIMP1 abrogates metastasis suppression. Lung cancer patients with metastasis and poor survival show low hRAB37 protein expression coinciding with low TIMP1 in tumours. Our findings identify hRAB37 as a novel metastasis suppressor Rab that functions through the TIMP1-MMP9 pathway and has significant prognostic power.


Subject(s)
Adenocarcinoma/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Matrix Metalloproteinase 9/genetics , Tissue Inhibitor of Metalloproteinase-1/genetics , rab GTP-Binding Proteins/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Animals , COS Cells , Cell Line, Tumor , Cell Movement , Chlorocebus aethiops , Epithelial Cells/metabolism , Epithelial Cells/pathology , Exocytosis/genetics , Humans , Injections, Intravenous , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Signal Transduction , Survival Analysis , Tail , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transport Vesicles/metabolism , rab GTP-Binding Proteins/metabolism
8.
Pflugers Arch ; 466(6): 1199-209, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24619480

ABSTRACT

The pathophysiology of heart failure (HF) is diverse, owing to multiple etiologies and aberrations in a number of cellular processes. Therefore, it is essential to understand how defects in the molecular pathways that mediate cellular responses to internal and external stressors function as a system to drive the HF phenotype. Mass spectrometry (MS)-based proteomics strategies have great potential for advancing our understanding of disease mechanisms at the systems level because proteins are the effector molecules for all cell functions and, thus, are directly responsible for determining cell phenotype. Two MS-based proteomics strategies exist: peptide-based bottom-up and protein-based top-down proteomics--each with its own unique strengths and weaknesses for interrogating the proteome. In this review, we will discuss the advantages and disadvantages of bottom-up and top-down MS for protein identification, quantification, and analysis of post-translational modifications, as well as highlight how both of these strategies have contributed to our understanding of the molecular and cellular mechanisms underlying HF. Additionally, the challenges associated with both proteomics approaches will be discussed and insights will be offered regarding the future of MS-based proteomics in HF research.


Subject(s)
Heart Failure/metabolism , Protein Processing, Post-Translational , Proteome/metabolism , Proteomics/methods , Amino Acid Sequence , Animals , Humans , Mass Spectrometry/methods , Molecular Sequence Data , Proteome/chemistry
9.
Biochim Biophys Acta ; 1834(11): 2285-92, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23376430

ABSTRACT

Secreted proteins, collectively referred to as the secretome, were suggested as valuable biomarkers in disease diagnosis and prognosis. However, some secreted proteins from cell cultures are difficult to detect because of their intrinsically low abundance; they are frequently masked by the released proteins from lysed cells and the substantial amounts of serum proteins used in culture medium. The hollow fiber culture (HFC) system is a commercially available system composed of small fibers sealed in a cartridge shell; cells grow on the outside of the fiber. Recently, because this system can help cells grow at a high density, it has been developed and applied in a novel analytical platform for cell secretome collection in cancer biomarker discovery. This article focuses on the advantages of the HFC system, including the effectiveness of the system for collection of secretomes, and reviews the process of cell secretome collection by the HFC system and proteomic approaches to discover cancer biomarkers. The HFC system not only provides a high-density three-dimensional (3D) cell culture system to mimic tumor growth conditions in vivo but can also accommodate numerous cells in a small volume, allowing secreted proteins to be accumulated and concentrated. In addition, cell lysis rates can be greatly reduced, decreasing the amount of contamination by abundant cytosolic proteins from lysed cells. Therefore, the HFC system is useful for preparing a wide range of proteins from cell secretomes and provides an effective method for collecting higher amounts of secreted proteins from cancer cells. This article is part of a Special Issue entitled: An Updated Secretome.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Culture Techniques/instrumentation , Neoplasms/diagnosis , Neoplasms/metabolism , Proteome/metabolism , Proteomics/instrumentation , Animals , Biomarkers, Tumor/analysis , Equipment Design , Humans , Proteome/analysis , Secretory Pathway
10.
J Proteome Res ; 11(11): 5167-85, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-22985211

ABSTRACT

As the leading cause of cancer death worldwide, lung cancer lacks effective diagnosis tools and treatments to prevent its metastasis. Fortunately, secretome has clinical usages as biomarkers and protein drugs. To discover the secretome that influences lung adenocarcinoma metastasis, the hollow fiber culture (HFC) system was used along with label-free proteomics approach to analyze cell secretomes between CL1-0 and CL1-5 cell lines, which exhibit low and high metastatic potentials. Among the 703 proteins quantified, 50 possessed different levels between CL1-0 and CL1-5. PARK7 was a primary focus because of the lack of research involving lung adenocarcinoma. The cell proliferation, migration, and invasion properties of CL1-0, CL1-5, and A549 cells were significantly diminished when the expression of their PARK7 proteins was reduced. Conversely, these functions were promoted when PARK7 was overexpressed in CL1-0. In clinical expression, PARK7 levels within tissue specimens and plasma samples were significantly higher in the cancer group. This represents the first time the HFC system has been used with label-free quantification to discern the elements of metastasis in lung adenocarcinoma cell secretomes. Likewise, PARK7 has never been researched for its role in promoting lung adenocarcinoma progression.


Subject(s)
Adenocarcinoma/metabolism , Cell Proliferation , Intracellular Signaling Peptides and Proteins/physiology , Lung Neoplasms/metabolism , Oncogene Proteins/physiology , Proteomics , Adenocarcinoma/pathology , Blotting, Western , Cell Line, Tumor , Culture Media, Serum-Free , Enzyme-Linked Immunosorbent Assay , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/pathology , Neoplasm Invasiveness , Neoplasm Metastasis , Oncogene Proteins/genetics , Protein Deglycase DJ-1 , Tandem Mass Spectrometry , Tissue Array Analysis
11.
Mol Cell Proteomics ; 11(11): 1320-39, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22896658

ABSTRACT

Metastasis is a major obstacle that must be overcome for the successful treatment of lung cancer. Proteins secreted by cancer cells may facilitate the progression of metastasis, particularly within the phases of migration and invasion. To discover metastasis-promoting secretory proteins within cancer cells, we used the label-free quantitative proteomics approach and compared the secretomes from the lung adenocarcinoma cell lines CL1-0 and CL1-5, which exhibit low and high metastatic properties, respectively. By employing quantitative analyses, we identified 660 proteins, 68 of which were considered to be expressed at different levels between the two cell lines. High levels of A1AT were secreted by CL1-5, and the roles of A1AT in the influence of lung adenocarcinoma metastasis were investigated. Molecular and pathological confirmation demonstrated that altered expression of A1AT correlates with the metastatic potential of lung adenocarcinoma. The migration and invasion properties of CL1-5 cells were significantly diminished by reducing the expression and secretion of their A1AT proteins. Conversely, the migration and invasion properties of CL1-0 cells were significantly increased through the overexpression and secretion of A1AT proteins. Furthermore, the assembly levels of the metastasis-promoting pericellular fibronectin (FN1), which facilitates colonization of lung capillary endothelia by adhering to the cell surface receptor dipeptidyl peptidase IV (DPP IV), were higher on the surfaces of suspended CL1-5 cells than on those of the CL1-0 cells. This discovery reflects previous findings in breast cancer. In line with this finding, FN1 assembly and the lung colonization of suspended CL1-5 cells were inhibited when endogenous A1AT protein was knocked down using siRNA. The major thrust of this study is to demonstrate the effects of coupling the label-free proteomics strategy with the secretomes of cancer cells that differentially exhibit invasive and metastatic properties. This provides a new opportunity for the effective identification of metastasis-associated proteins that are secreted by cancer cells and promote experimental metastasis.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Cell Movement , Fibronectins/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung/pathology , Neoplasm Proteins/metabolism , alpha 1-Antitrypsin/metabolism , Adenocarcinoma of Lung , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Humans , Immunohistochemistry , Lung/metabolism , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Polymerization/drug effects , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...