Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
Cell Rep Med ; 5(5): 101547, 2024 May 21.
Article En | MEDLINE | ID: mdl-38703764

Non-clear cell renal cell carcinomas (non-ccRCCs) encompass diverse malignant and benign tumors. Refinement of differential diagnosis biomarkers, markers for early prognosis of aggressive disease, and therapeutic targets to complement immunotherapy are current clinical needs. Multi-omics analyses of 48 non-ccRCCs compared with 103 ccRCCs reveal proteogenomic, phosphorylation, glycosylation, and metabolic aberrations in RCC subtypes. RCCs with high genome instability display overexpression of IGF2BP3 and PYCR1. Integration of single-cell and bulk transcriptome data predicts diverse cell-of-origin and clarifies RCC subtype-specific proteogenomic signatures. Expression of biomarkers MAPRE3, ADGRF5, and GPNMB differentiates renal oncocytoma from chromophobe RCC, and PIGR and SOSTDC1 distinguish papillary RCC from MTSCC. This study expands our knowledge of proteogenomic signatures, biomarkers, and potential therapeutic targets in non-ccRCC.


Biomarkers, Tumor , Carcinoma, Renal Cell , Kidney Neoplasms , Proteogenomics , Humans , Proteogenomics/methods , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/metabolism , Transcriptome/genetics , Male , Female , Middle Aged , Gene Expression Regulation, Neoplastic
2.
Cell Rep Med ; 4(9): 101173, 2023 09 19.
Article En | MEDLINE | ID: mdl-37582371

We introduce a pioneering approach that integrates pathology imaging with transcriptomics and proteomics to identify predictive histology features associated with critical clinical outcomes in cancer. We utilize 2,755 H&E-stained histopathological slides from 657 patients across 6 cancer types from CPTAC. Our models effectively recapitulate distinctions readily made by human pathologists: tumor vs. normal (AUROC = 0.995) and tissue-of-origin (AUROC = 0.979). We further investigate predictive power on tasks not normally performed from H&E alone, including TP53 prediction and pathologic stage. Importantly, we describe predictive morphologies not previously utilized in a clinical setting. The incorporation of transcriptomics and proteomics identifies pathway-level signatures and cellular processes driving predictive histology features. Model generalizability and interpretability is confirmed using TCGA. We propose a classification system for these tasks, and suggest potential clinical applications for this integrated human and machine learning approach. A publicly available web-based platform implements these models.


Deep Learning , Neoplasms , Proteogenomics , Humans , Neoplasms/genetics , Proteomics , Machine Learning
3.
Cancer Cell ; 41(9): 1586-1605.e15, 2023 09 11.
Article En | MEDLINE | ID: mdl-37567170

We characterized a prospective endometrial carcinoma (EC) cohort containing 138 tumors and 20 enriched normal tissues using 10 different omics platforms. Targeted quantitation of two peptides can predict antigen processing and presentation machinery activity, and may inform patient selection for immunotherapy. Association analysis between MYC activity and metformin treatment in both patients and cell lines suggests a potential role for metformin treatment in non-diabetic patients with elevated MYC activity. PIK3R1 in-frame indels are associated with elevated AKT phosphorylation and increased sensitivity to AKT inhibitors. CTNNB1 hotspot mutations are concentrated near phosphorylation sites mediating pS45-induced degradation of ß-catenin, which may render Wnt-FZD antagonists ineffective. Deep learning accurately predicts EC subtypes and mutations from histopathology images, which may be useful for rapid diagnosis. Overall, this study identified molecular and imaging markers that can be further investigated to guide patient stratification for more precise treatment of EC.


Endometrial Neoplasms , Metformin , Proteogenomics , Female , Humans , Proto-Oncogene Proteins c-akt/genetics , Prospective Studies , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , beta Catenin/genetics , beta Catenin/metabolism , Metformin/pharmacology
4.
Cancers (Basel) ; 13(10)2021 May 12.
Article En | MEDLINE | ID: mdl-34066104

Therapeutic guidance in non-small cell lung cancer (NSCLC) tumors that are positive for anaplastic lymphoma kinase (ALK) fluorescent in situ hybridization (FISH), but negative for ALK immunohistochemistry, is still challenging. Parallel routine screening of 4588 NSCLC cases identified 22 discordant cases. We rechecked these samples using ALK antibodies and selected reaction monitoring (SRM) quantitative multiplexed proteomics screening multiple protein targets, including ALK and MET for the ALK tyrosine kinase inhibitor (TKI), and FR-alpha, hENT1, RRM1, TUBB3, ERCC1, and XRCC1 for chemotherapy. The presence of ALK (31.8%), MET (36.4%), FR-alpha (72.7%), hENT1 (18.2%), RRM1 (31.8%), TUBB3 (72.9%), ERCC1 (4.5%), and a low level of XRCC1 (54.4%) correlated with clinical outcomes. SRM was more sensitive than the ALK D5F3 assay. Among the eight cases receiving ALK TKI, four cases with ALK or MET detected by SRM had complete or partial responses, whereas four cases without ALK or MET showed progression. Twenty-seven treatment outcomes from 20 cases were assessed and cases expressing more than half of the specific predictive proteins were sensitive to matching therapeutic agents and showed longer progression-free survival than the other cases (p < 0.001). SRM showed a potential role in therapeutic decision making in NSCLC patients with ambiguous ALK test results.

5.
J Cardiovasc Magn Reson ; 23(1): 76, 2021 06 17.
Article En | MEDLINE | ID: mdl-34134713

BACKGROUND: Chemotherapy-induced cardiotoxicity is a well-recognized adverse effect of chemotherapy. Quantitative T1-mapping cardiovascular magnetic resonance (CMR) is useful for detecting subclinical myocardial changes in anthracycline-induced cardiotoxicity. The aim of the present study was to histopathologically validate the T1 and T2 mapping parameters for the evaluation of diffuse myocardial changes in rat models of cardiotoxicity. METHODS: Rat models of cardiotoxicity were generated by injecting rats with doxorubicin (1 mg/kg, twice a week). CMR was performed with a 9.4 T ultrahigh-field scanner using cine, pre-T1, post-T1 and T2 mapping sequences to evaluate the left ventricular ejection fraction (LVEF), native T1, T2, and extracellular volume fraction (ECV). Histopathological examinations were performed and the association of histopathological changes with CMR parameters was assessed. RESULTS: Five control rats and 36 doxorubicin-treated rats were included and classified into treatment periods. In the doxorubicin-treated rats, the LVEF significantly decreased after 12 weeks of treatment (control vs. 12-week treated: 73 ± 4% vs. 59 ± 9%, P = 0.01).  Increased native T1 and ECV were observed after 6 weeks of treatment (control vs. 6-week treated: 1148 ± 58 ms, 14.3 ± 1% vs. 1320 ± 56 ms, 20.3 ± 3%; P = 0.005, < 0.05, respectively). T2 values also increased by six weeks of treatment (control vs. 6-week treated: 16.3 ± 2 ms vs. 10.3 ± 1 ms, P < 0.05). The main histopathological features were myocardial injury, interstitial fibrosis, inflammation, and edema. The mean vacuolar change (%), fibrosis (%), and inflammation score were significantly higher in 6-week treated rats than in the controls (P = 0.03, 0.03, 0.02, respectively). In the univariable analysis, vacuolar change showed the highest correlation with native T1 value (R = 0.60, P < 0.001), and fibrosis showed the highest correlation with ECV value (R = 0.78, P < 0.001). In the multiple linear regression analysis model, vacuolar change was a significant factor for change in native T1 (P = 0.01), and vacuolar change and fibrosis were significant factors for change in ECV (P = 0.006, P < 0.001, respectively) by adding other histopathological parameters (i.e., inflammation and edema scores) CONCLUSIONS: Quantitative T1 and T2 mapping CMR is a useful non-invasive tool reflecting subclinical histopathological changes in anthracycline-induced cardiotoxicity.


Anthracyclines , Cardiotoxicity , Animals , Cardiotoxicity/pathology , Fibrosis , Magnetic Resonance Imaging, Cine , Magnetic Resonance Spectroscopy , Myocardium/pathology , Predictive Value of Tests , Rats , Stroke Volume , Ventricular Function, Left
6.
Cancer Cell ; 39(3): 361-379.e16, 2021 03 08.
Article En | MEDLINE | ID: mdl-33417831

We present a proteogenomic study of 108 human papilloma virus (HPV)-negative head and neck squamous cell carcinomas (HNSCCs). Proteomic analysis systematically catalogs HNSCC-associated proteins and phosphosites, prioritizes copy number drivers, and highlights an oncogenic role for RNA processing genes. Proteomic investigation of mutual exclusivity between FAT1 truncating mutations and 11q13.3 amplifications reveals dysregulated actin dynamics as a common functional consequence. Phosphoproteomics characterizes two modes of EGFR activation, suggesting a new strategy to stratify HNSCCs based on EGFR ligand abundance for effective treatment with inhibitory EGFR monoclonal antibodies. Widespread deletion of immune modulatory genes accounts for low immune infiltration in immune-cold tumors, whereas concordant upregulation of multiple immune checkpoint proteins may underlie resistance to anti-programmed cell death protein 1 monotherapy in immune-hot tumors. Multi-omic analysis identifies three molecular subtypes with high potential for treatment with CDK inhibitors, anti-EGFR antibody therapy, and immunotherapy, respectively. Altogether, proteogenomics provides a systematic framework to inform HNSCC biology and treatment.


Antineoplastic Agents, Immunological/therapeutic use , Papillomavirus Infections/genetics , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics , Adult , Aged , Aged, 80 and over , ErbB Receptors/genetics , Female , Humans , Immunotherapy/methods , Male , Middle Aged , Papillomavirus Infections/drug therapy , Papillomavirus Infections/virology , Proteogenomics/methods , Proteomics/methods , Young Adult
7.
BMC Cardiovasc Disord ; 20(1): 264, 2020 06 03.
Article En | MEDLINE | ID: mdl-32493217

BACKGROUND: Cardiovascular disease is second only to cancer recurrence as a determinant of lifespan in cancer survivors, and cancer therapy-related cardiac dysfunction is a clinically important risk factor. We aim to investigate the use of cardiac magnetic resonance imaging (MRI) to evaluate early tissue changes and perform functional assessment of chemo- and radiation-induced cardiotoxicity and to identify MRI prognostic indicators of cardiotoxicity in breast cancer patients. METHODS: A 3-min cardiac imaging protocol will be added to the breast MRI examination to diagnose cardiotoxicity in breast cancer patients. Standardized MRI-based evaluation of breast cancer and the left ventricular myocardium will be performed at baseline and at 3, 6, and 12 months and 2 years or more after cancer treatment. We will analyze both ventricular volume and ejection fraction (EF), strain of left ventricle (LV), native T1, extracellular volume fraction (ECV), and T2 values acquired in the mid LV. DISCUSSION: The primary result of this study will be the comparison of the prognostic value of MRI parameters (native T1, ECV, both ventricular systolic function and LV strain) for cardiotoxicity. The endpoint is defined as the occurrence of a major adverse cardiac event (MACE). The secondary outcome will be an assessment of the temporal relationships between contractile dysfunction and microstructural injury over 4 years using MRI. This study will assess the usefulness of quantitative MRI to diagnose cardiotoxicity and will clarify the temporal relationships between contractile dysfunction and microstructural injury of the LV myocardium using MRI during breast cancer treatment. TRIAL REGISTRATION: The protocol was registered at clinicaltrials.gov (Clinical trial no. NCT03301389) on October 4, 2017.


Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/therapy , Heart Diseases/diagnostic imaging , Magnetic Resonance Imaging, Cine , Radiation Injuries/diagnostic imaging , Stroke Volume/drug effects , Ventricular Function, Left/drug effects , Cardiotoxicity , Clinical Trials as Topic , Female , Heart Disease Risk Factors , Heart Diseases/chemically induced , Heart Diseases/physiopathology , Humans , Predictive Value of Tests , Prospective Studies , Radiation Injuries/etiology , Radiotherapy/adverse effects , Risk Assessment , Seoul , Time Factors
8.
ACS Biomater Sci Eng ; 6(8): 4390-4396, 2020 08 10.
Article En | MEDLINE | ID: mdl-33455188

A hybrid composite of silver nanoparticles (AgNPs) and porous silicon microparticles (pSiMPs) was developed and applied for the computed tomography (CT) scanning of the lungs as an image-guided localization agent. We confirmed the grafting of AgNPs on oxidized pSiMPs template using various analytical equipment, including a scanning electron microscope (SEM), Fourier transform infrared (FTIR) spectroscopy, X-ray diffraction (XRD), and energy-dispersive X-ray spectroscopy (EDS). The hybrid composite showed a high CT contrast intensity (>1000 HU) that enabled us to produce and view images of the lungs. In addition, it showed the ability to maintain a strong CT signal at the injected area of the rabbit's lungs, up to an hour, without spreading. The lack of toxicity and immune response indicated that the composite could be fully utilized as a new image-guided localization agent of CT scans for lung cancer surgery.


Metal Nanoparticles , Silver , Animals , Lung/diagnostic imaging , Porosity , Rabbits , Silicon , Tomography , Tomography, X-Ray Computed
9.
Clin Chem ; 62(1): 252-61, 2016 Jan.
Article En | MEDLINE | ID: mdl-26585927

BACKGROUND: Crizotinib has antitumor activity in ALK (anaplastic lymphoma receptor tyrosine kinase)-rearranged non-small cell lung cancer (NSCLC). The current diagnostic test for ALK rearrangement is breakapart fluorescence in situ hybridization (FISH), but FISH has low throughput and is not always reflective of protein concentrations. The emergence of multiple clinically relevant biomarkers in NSCLC necessitates efficient testing of scarce tissue samples. We developed an anaplastic lymphoma kinase (ALK) protein assay that uses multiplexed selected reaction monitoring (SRM) to quantify absolute amounts of ALK in formalin-fixed paraffin-embedded (FFPE) tumor tissue. METHODS: After validation in formalin-fixed cell lines, the SRM assay was used to quantify concentrations of ALK in 18 FFPE NSCLC samples that had been tested for ALK by FISH and immunohistochemistry. Results were correlated with patient response to crizotinib. RESULTS: We detected ALK in 11 of 14 NSCLC samples with known ALK rearrangements by FISH. Absolute ALK concentrations correlated with clinical response in 5 of 8 patients treated with crizotinib. The SRM assay did not detect ALK in 3 FISH-positive patients who had not responded to crizotinib. In 1 of these cases, DNA sequencing revealed a point mutation that predicts a nonfunctional ALK fusion protein. The SRM assay did not detect ALK in any tumor tissue with a negative ALK status by FISH or immunohistochemistry. CONCLUSIONS: ALK concentrations measured by SRM correlate with crizotinib response in NSCLC patients. The ALK SRM proteomic assay, which may be multiplexed with other clinically relevant proteins, allows for rapid identification of patients potentially eligible for targeted therapies.


Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Gene Expression Regulation, Neoplastic , Lung Neoplasms/drug therapy , Pyrazoles/therapeutic use , Pyridines/therapeutic use , Receptor Protein-Tyrosine Kinases/analysis , Receptor Protein-Tyrosine Kinases/genetics , Anaplastic Lymphoma Kinase , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Crizotinib , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Pyrazoles/pharmacology , Pyridines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors
10.
Mol Cell Proteomics ; 14(10): 2661-81, 2015 Oct.
Article En | MEDLINE | ID: mdl-26199343

Osteoclasts are monocyte-derived multinuclear cells that directly attach to and resorb bone. Sphingosine-1-phosphate (S1P)(1) regulates bone resorption by functioning as both a chemoattractant and chemorepellent of osteoclast precursors through two G-protein coupled receptors that antagonize each other in an S1P-concentration-dependent manner. To quantitatively explore the behavior of this chemosensing pathway, we applied targeted proteomics, transcriptomics, and rule-based pathway modeling using the Simmune toolset. RAW264.7 cells (a mouse monocyte/macrophage cell line) were used as model osteoclast precursors, RNA-seq was used to identify expressed target proteins, and selected reaction monitoring (SRM) mass spectrometry using internal peptide standards was used to perform absolute abundance measurements of pathway proteins. The resulting transcript and protein abundance values were strongly correlated. Measured protein abundance values, used as simulation input parameters, led to in silico pathway behavior matching in vitro measurements. Moreover, once model parameters were established, even simulated responses toward stimuli that were not used for parameterization were consistent with experimental findings. These findings demonstrate the feasibility and value of combining targeted mass spectrometry with pathway modeling for advancing biological insight.


Chemotaxis/physiology , Lysophospholipids/metabolism , Macrophages/metabolism , Proteomics , Sphingosine/analogs & derivatives , Animals , Cell Line , Macrophages/physiology , Mice , Sequence Analysis, RNA , Signal Transduction , Sphingosine/metabolism
11.
Mol Cell Proteomics ; 13(10): 2687-704, 2014 Oct.
Article En | MEDLINE | ID: mdl-25044017

In addition to forming macrophages and dendritic cells, monocytes in adult peripheral blood retain the ability to develop into osteoclasts, mature bone-resorbing cells. The extensive morphological and functional transformations that occur during osteoclast differentiation require substantial reprogramming of gene and protein expression. Here we employ -omic-scale technologies to examine in detail the molecular changes at discrete developmental stages in this process (precursor cells, intermediate osteoclasts, and multinuclear osteoclasts), quantitatively comparing their transcriptomes and proteomes. The data have been deposited to the ProteomeXchange with identifier PXD000471. Our analysis identified mitochondrial changes, along with several alterations in signaling pathways, as central to the development of mature osteoclasts, while also confirming changes in pathways previously implicated in osteoclast biology. In particular, changes in the expression of proteins involved in metabolism and redirection of energy flow from basic cellular function toward bone resorption appeared to play a key role in the switch from monocytic immune system function to specialized bone-turnover function. These findings provide new insight into the differentiation program involved in the generation of functional osteoclasts.


Cellular Reprogramming , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Osteoclasts/metabolism , Proteomics/methods , Animals , Cell Line , Mice , Mitochondria/genetics , Mitochondria/metabolism , Osteoclasts/cytology , Proteome/analysis , RANK Ligand/pharmacology , Signal Transduction
12.
Cell ; 150(2): 317-26, 2012 Jul 20.
Article En | MEDLINE | ID: mdl-22817894

In eukaryotes, DNA is packaged into chromatin by canonical histone proteins. The specialized histone H3 variant CENP-A provides an epigenetic and structural basis for chromosome segregation by replacing H3 at centromeres. Unlike exclusively octameric canonical H3 nucleosomes, CENP-A nucleosomes have been shown to exist as octamers, hexamers, and tetramers. An intriguing possibility reconciling these observations is that CENP-A nucleosomes cycle between octamers and tetramers in vivo. We tested this hypothesis by tracking CENP-A nucleosomal components, structure, chromatin folding, and covalent modifications across the human cell cycle. We report that CENP-A nucleosomes alter from tetramers to octamers before replication and revert to tetramers after replication. These structural transitions are accompanied by reversible chaperone binding, chromatin fiber folding changes, and previously undescribed modifications within the histone fold domains of CENP-A and H4. Our results reveal a cyclical nature to CENP-A nucleosome structure and have implications for the maintenance of epigenetic memory after centromere replication.


Autoantigens/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Nucleosomes/metabolism , Autoantigens/chemistry , Cell Cycle , Centromere/metabolism , Centromere Protein A , Chromosomal Proteins, Non-Histone/chemistry , DNA Replication , DNA-Binding Proteins/metabolism , HEK293 Cells , HeLa Cells , Histones/chemistry , Histones/metabolism , Humans , Models, Molecular , Protein Structure, Tertiary
13.
Expert Rev Proteomics ; 9(3): 337-45, 2012 Jun.
Article En | MEDLINE | ID: mdl-22809211

Studies of the cell secretome have greatly increased in recent years owing to improvements in proteomic platforms, mass spectrometry instrumentation and to the increased interaction between analytical chemists, biologists and clinicians. Several secretome studies have been implemented in different areas of research, leading to the generation of a valuable secretome catalogs. Secreted proteins continue to be an important source of biomarkers and therapeutic target discovery and are equally valuable in the field of microbiology. Several discoveries have been achieved in vitro using cell culture systems, ex vivo using human tissue specimens and in vivo using animal models. In this review, some of the most recent advances in secretome studies and the fields that have benefited the most from this evolving technology are highlighted.


Proteins/analysis , Proteins/metabolism , Proteomics/methods , Amniotic Fluid/chemistry , Biomarkers, Tumor/analysis , Bodily Secretions/chemistry , Body Fluids/chemistry , Exosomes/chemistry , Humans , Mass Spectrometry/methods , Neoplasms/metabolism
14.
Invest Ophthalmol Vis Sci ; 51(7): 3379-86, 2010 Jul.
Article En | MEDLINE | ID: mdl-20207970

PURPOSE. To define the role of the serine protease HTRA1 in age-related macular degeneration (AMD) by examining its expression level and identifying its potential substrates in the context of primary RPE cell extracellular milieu. METHODS. Primary RPE cell cultures were established from human donor eyes and screened for CFH, ARMS2, and HTRA1 risk genotypes by using an allele-discrimination assay. HTRA1 expression in genotyped RPE cells was determined by using real-time PCR and quantitative proteomics. Potential HTRA1 substrates were identified by incubating RPE-conditioned medium with or without human recombinant HTRA1. Selectively cleaved proteins were quantified by using the differential stable isotope labeling by amino acids in cell culture (SILAC) strategy. RESULTS. HTRA1 mRNA levels were threefold higher in primary RPE cells homozygous for the HTRA1 promoter risk allele than in RPE cells with the wild-type allele, which translated into a twofold increase in HTRA1 secretion by RPE cells with the risk genotype. A total of 196 extracellular proteins were identified in the RPE secretome, and only 8 were found to be selectively cleaved by the human recombinant HTRA1. These include fibromodulin with 90% cleavage, clusterin (50%), ADAM9 (54%), vitronectin (54%), and alpha2-macroglobulin (55%), as well as some cell surface proteins including talin-1 (21%), fascin (40%), and chloride intracellular channel protein 1 (51%). CONCLUSIONS. Recombinant HTRA1 cleaves RPE-secreted proteins involved in regulation of the complement pathway (clusterin, vitronectin, and fibromodulin) and of amyloid deposition (clusterin, alpha2-macroglobulin, and ADAM9). These findings suggest a link between HTRA1, complement regulation, and amyloid deposition in AMD pathogenesis.


Amyloid/metabolism , Complement Pathway, Classical/physiology , Macular Degeneration/enzymology , Retinal Pigment Epithelium/enzymology , Serine Endopeptidases/physiology , ADAM Proteins/metabolism , Aged , Blotting, Western , Cells, Cultured , Chromatography, High Pressure Liquid , Clusterin/metabolism , Complement Factor H/genetics , Electrophoresis, Polyacrylamide Gel , Extracellular Matrix Proteins/metabolism , Fibromodulin , Genotype , High-Temperature Requirement A Serine Peptidase 1 , Humans , Membrane Proteins/metabolism , Proteins/genetics , Proteoglycans/metabolism , Proteomics , RNA, Messenger/metabolism , Recombinant Proteins , Reverse Transcriptase Polymerase Chain Reaction , Substrate Specificity , Tandem Mass Spectrometry , Vitronectin/metabolism , alpha-Macroglobulins/metabolism
15.
Mol Vis ; 14: 2292-303, 2008.
Article En | MEDLINE | ID: mdl-19093006

PURPOSE: To identify cytokine-induced changes in the secretome of human retinal pigment epithelial (RPE) cells and their potential implication in age-related macular degeneration pathogenesis. METHODS: Stable isotope labeling by amino in cell culture (SILAC) was used in combination with liquid chromatography tandem mass spectrometry (LC-MS/MS) to measure differential protein secretion from tumor necrosis factor-alpha (TNF-alpha) treated ARPE-19 versus untreated ARPE-19 cells. Typically, one set of cells was subcultured in a medium in which Arg and Lys were replaced by (13)C(6)-Arg and (15)N(2,) (13)C(6)-Lys while the other set of cells was grown in unlabeled medium. The fully labeled cells were then treated with TNF-alpha, while unlabeled cells were left untreated. Spent media from both treated and untreated cells were collected, mixed at 1:1 ratio, and processed for LC-MS/MS analysis. Labeled and unlabeled peptide pairs were identified and their intensities were used to determine protein ratios in TNF-alpha treated cells versus untreated cells. To validate the data, we performed a reverse experiment in which unlabeled cells were treated with TNF-alpha while labeled cells were kept untreated. RESULTS: A total of 146 proteins were identified as putatively secreted proteins in the spent medium of ARPE-19 cells and only six among these were differentially secreted following TNF-alpha treatment. Secretion of complement 3 and sulfhydryl oxidase-1 was increased by twofold, fibronectin by 1.7 fold, plasminogen activator inhibitor 1 by 1.9 fold and syndecan-4 by 4.35 fold while secretion of trans-golgi network protein-2 was decreased by twofold. CONCLUSIONS: TNF-alpha modulates secretion of specific proteins in ARPE-19 cells. These proteins are involved in pathways relevant to AMD pathogenesis (e.g., extracellular matrix remodeling, complement pathway, and angiogenesis).


Eye Proteins/metabolism , Pigment Epithelium of Eye/cytology , Pigment Epithelium of Eye/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Cell Line , Cell Survival/drug effects , Humans , Pigment Epithelium of Eye/drug effects
16.
J Proteome Res ; 5(10): 2599-610, 2006 Oct.
Article En | MEDLINE | ID: mdl-17022631

Age-related macular degeneration (AMD) is characterized by progressive loss of central vision, which is attributed to abnormal accumulation of macular deposits called "drusen" at the interface between the basal surface of the retinal pigment epithelium (RPE) and Bruch's membrane. In the most severe cases, drusen deposits are accompanied by the growth of new blood vessels that breach the RPE layer and invade photoreceptors. In this study, we hypothesized that RPE secreted proteins are responsible for drusen formation and choroidal neovascularization. We used stable isotope labeling by amino acids in cell culture (SILAC) in combination with LC-MS/MS analysis and ZoomQuant quantification to assess differential protein secretion by RPE cell cultures prepared from human autopsy eyes of AMD donors (diagnosed by histological examinations of the macula and genotyped for the Y402H-complement factor H variant) and age-matched healthy control donors. In general, RPE cells were found to secrete a variety of extracellular matrix proteins, complement factors, and protease inhibitors that have been reported to be major constituents of drusen (hallmark deposits in AMD). Interestingly, RPE cells from AMD donors secreted 2 to 3-fold more galectin 3 binding protein, fibronectin, clusterin, matrix metalloproteinase-2 and pigment epithelium derived factor than RPE cells from age-matched healthy donors. Conversely, secreted protein acidic and rich in cysteine (SPARC) was found to be down regulated by 2-fold in AMD RPE cells versus healthy RPE cells. Ingenuity pathway analysis grouped these differentially secreted proteins into two groups; those involved in tissue development and angiogenesis and those involved in complement regulation and protein aggregation such as clusterin. Overall, these data strongly suggest that RPE cells are involved in the biogenesis of drusen and the pathology of AMD.


Macular Degeneration/metabolism , Pigment Epithelium of Eye/metabolism , Proteins/analysis , Proteome/analysis , Tissue Donors , Aged , Cells, Cultured , Chromatography, Liquid , Humans , Macula Lutea/ultrastructure , Macular Degeneration/pathology , Mass Spectrometry , Microscopy, Electron , Pigment Epithelium of Eye/ultrastructure , Protein Transport , Proteins/metabolism , Proteome/metabolism , Retinal Drusen/etiology , Retinal Drusen/metabolism , Retinal Drusen/pathology
...