Elsevier

Journal of Proteomics

Volume 75, Issue 8, 18 April 2012, Pages 2384-2395
Journal of Proteomics

Label-free protein profiling of formalin-fixed paraffin-embedded (FFPE) heart tissue reveals immediate mitochondrial impairment after ionising radiation

https://doi.org/10.1016/j.jprot.2012.02.019Get rights and content

Abstract

Qualitative proteome profiling of formalin-fixed, paraffin-embedded (FFPE) tissue is advancing the field of clinical proteomics. However, quantitative proteome analysis of FFPE tissue is hampered by the lack of an efficient labelling method. The usage of conventional protein labelling on FFPE tissue has turned out to be inefficient. Classical labelling targets lysine residues that are blocked by the formalin treatment. The aim of this study was to establish a quantitative proteomics analysis of FFPE tissue by combining the label-free approach with optimised protein extraction and separation conditions.

As a model system we used FFPE heart tissue of control and exposed C57BL/6 mice after total body irradiation using a gamma ray dose of 3 gray. We identified 32 deregulated proteins (p  0.05) in irradiated hearts 24 h after the exposure. The proteomics data were further evaluated and validated by bioinformatics and immunoblotting investigation. In good agreement with our previous results using fresh-frozen tissue, the analysis indicated radiation-induced alterations in three main biological pathways: respiratory chain, lipid metabolism and pyruvate metabolism. The label-free approach enables the quantitative measurement of radiation-induced alterations in FFPE tissue and facilitates retrospective biomarker identification using clinical archives.

Graphical abstract

Highlights

► Label-free method is suitable for quantitative proteome analysis of FFPE tissue. ► Ionising radiation (IR) induces changes in three main biological pathways. ► IR causes down-regulation of mitochondrial complex proteins. ► Fatty acid metabolism and citric acid cycle were impaired by IR. ► Radiation-induced alteration in the glucose oxidative pathway was observed.

Introduction

For decades, pathologists have been using formalin-fixed, paraffin-embedded (FFPE) tissue for histological analysis due to its excellent performance and suitability for long-term storage. Clinical archives including data on diagnosis and outcome may provide information on biological pathways and cellular processes leading to disease, provided that suitable molecular technologies are available [1], [2], [3].

Proteomics analysis using FFPE material as an alternative to fresh-frozen tissue has recently been investigated [4], [5], [6], [7]. Different methods of extraction and separation of proteins from FFPE samples have been established [8], [9], [10], [11]. However, quantitative proteomic studies on archival material have been considered as an almost impossible task, primarily due to the harsh and irreversible fixation procedures and loss of integrity during prolonged storage. The formaldehyde fixation leads to a methylol modification, tagged mainly on lysine residues [11], [12]. Most chemical labels used in quantitative proteomics also target lysine residues, leading to inefficient labelling of FFPE material [13], [14], [15].

Recently, non-labelling approaches have been suggested as an alternative for quantification of FFPE proteome profiles [16], [17]. The few quantitative proteomic studies published so far have been performed using tumour FFPE tissues where areas of interest are easily distinguished and dissected [18], [19], thereby reducing the complexity of the resultant proteome profile.

The aim of this study was to establish a quantitative proteomics work flow using the label-free approach [20] in a tissue where no discrete target area can be observed. We have shown previously that ionising radiation has both immediate and persistent effects on the murine cardiac proteome without causing any morphological changes in the heart [21], [22]. Therefore, we used the cardiac tissue of sham- and total body irradiated C57BL/6 mice as a model system. We applied the optimised protein extraction and separation conditions for FFPE tissue previously developed by us for qualitative proteomics [11]. The analysis indicated radiation-induced impairment of the cardiac mitochondrial proteome. The proteomics data were further evaluated and validated by bioinformatics and immunoblotting investigation.

Section snippets

Materials

Beta-octylglucoside, SDS, and ammonium bicarbonate were obtained from Sigma (St. Louis, MO); RapiGest from Waters (USA); acetone, acetonitrile, formic acid, and trifluoroacetic acid (TFA) from Roth (Karlsuhe, Germany); dithiothreitol (DTT), iodoacetamide, tris-(hydroxymethyl) aminomethane (Tris) and sequencing grade trypsin were obtained from Promega (Madison, WI); cyano-4-hydroxycinnamic acid was obtained from Bruker Daltonik (Bremen, Germany). All solutions were prepared using HPLC grade

Protein quantification and identification

In order to evaluate the technical variability of mass spectrometry runs of label-free peptide quantifications of FFPE samples, we first analysed an aliquot of one control sample by repetitive LC-MS/MS runs using four replicates. After alignment of all peptide intensities and identification of respective proteins, the cumulative intensities of all proteins were plotted against their counterpart in the technical replicates (Supplementary Fig. 1). The resulting scatter plots showed that all four

Discussion

Recent epidemiological data on exposed individuals clearly show that ionising radiation is a risk factor for cardiovascular disease [28], [29], [30]. However, the molecular mechanisms underlying the development of radiation-induced heart disease are not well understood so far. Proteomics is a powerful technology that can provide novel information about such biological mechanisms. In this study, we performed a quantitative proteomic analysis using FFPE cardiac tissue from exposed and non-exposed

Conclusions

Our study shows that label-free proteomic approach enables to detect and quantify radiation-induced alterations in FFPE tissue. It indicates that archival tissue, that frequently is the only source of biomaterial available, plays a valuable role in retrospective molecular studies. This is particularly valid in cardiovascular diseases where additional collecting and sampling of fresh-frozen biomaterial is for ethical reasons cumbersome. We suggest that the three pathways found

Acknowledgements

This work was supported by a grant from the European Community's Seventh Framework Programme (EURATOM) contract no. 232628 (STORE). We acknowledge Dr. Steven E. Lipshultz for the valuable advice and discussions in the preparation of this manuscript. We thank Dr. Hakan Sarioglu, Sandra Helm and Waldemar Schneider for their technical assistance.

References (61)

  • P.M. Barger et al.

    PPAR signaling in the control of cardiac energy metabolism

    Trends Cardiovasc Med

    (2000)
  • L. Khouzami et al.

    Delayed cardiomyopathy in dystrophin deficient mdx mice relies on intrinsic glutathione resource

    Am J Pathol

    (2010)
  • D.R. Marenstein et al.

    Stimulation of human endonuclease III by Y box-binding protein 1 (DNA-binding protein B). Interaction between a base excision repair enzyme and a transcription factor

    J Biol Chem

    (2001)
  • A. Bieler et al.

    Impact of radiation therapy on the oncolytic adenovirus dl520: implications on the treatment of glioblastoma

    Radiother Oncol

    (2008)
  • S. Tapio et al.

    Molecular information obtained from radiobiological tissue archives: achievements of the past and visions of the future

    Radiat Environ Biophys

    (2008)
  • S. Tapio et al.

    Use of proteomics in radiobiological research: current state of the art

    Radiat Environ Biophys

    (2010)
  • S. Tapio et al.

    Progress in updating the European Radiobiology Archives

    Int J Radiat Biol

    (2008)
  • B.L. Hood et al.

    Mass spectrometric analysis of formalin-fixed paraffin-embedded tissue: unlocking the proteome within

    Proteomics

    (2006)
  • N.J. Nirmalan et al.

    Development and validation of a novel protein extraction methodology for quantitation of protein expression in formalin-fixed paraffin-embedded tissues using western blotting

    J Pathol

    (2009)
  • J. Nazarian et al.

    Protein profiling of formalin fixed paraffin embedded tissue: identification of potential biomarkers for pediatric brainstem glioma

    Proteomics Clin Appl

    (2008)
  • S.R. Shi et al.

    Protein extraction from formalin-fixed, paraffin-embedded tissue sections: quality evaluation by mass spectrometry

    J Histochem Cytochem

    (2006)
  • K.F. Becker et al.

    Quantitative protein analysis from formalin-fixed tissues: implications for translational clinical research and nanoscale molecular diagnosis

    J Pathol

    (2007)
  • O. Azimzadeh et al.

    Formalin-fixed paraffin-embedded (FFPE) proteome analysis using gel-free and gel-based proteomics

    J Proteome Res

    (2010)
  • Z. Xiao et al.

    Quantitative proteomic analysis of formalin-fixed and paraffin-embedded nasopharyngeal carcinoma using iTRAQ labeling, two-dimensional liquid chromatography, and tandem mass spectrometry

    J Histochem Cytochem

    (2010)
  • M.R. Jain et al.

    Quantitative proteomic analysis of formalin fixed paraffin embedded oral HPV lesions from HIV patients

    Open Proteomics J

    (2008)
  • E. Donadio et al.

    Evaluation of formalin-fixed paraffin-embedded tissues in the proteomic analysis of parathyroid glands

    Proteome Sci

    (2011)
  • P. Ostasiewicz et al.

    Proteome, phosphoproteome, and N-glycoproteome are quantitatively preserved in formalin-fixed paraffin-embedded tissue and analyzable by high-resolution mass spectrometry

    J Proteome Res

    (2010)
  • V. Patel et al.

    Proteomic analysis of laser-captured paraffin-embedded tissues: a molecular portrait of head and neck cancer progression

    Clin Cancer Res

    (2008)
  • O. Azimzadeh et al.

    Rapid proteomic remodeling of cardiac tissue caused by total body ionizing radiation

    Proteomics

    (2011)
  • Z. Barjaktarovic et al.

    Radiation–induced Signaling Results in Mitochondrial Impairment in Mouse Heart at 4 Weeks after Exposure to X-rays

    PLoS One

    (2011)
  • Cited by (33)

    • Proteomic developments in the analysis of formalin-fixed tissue

      2015, Biochimica et Biophysica Acta - Proteins and Proteomics
      Citation Excerpt :

      From these papers (excluding those that used a pre-fractionation step), the average number of unique protein identifications was 274 ± 222. In more recent times standalone Ion Traps have been replaced with more advanced instrumentation such as Ion Trap/Orbitrap hybrids, combining the sensitivity of the Ion Trap with the high resolution and mass accuracy of the Orbitrap, as seen in 20 papers since 2010, summarized in Table 2 [31,33,39–41,43,53,61,65,66,80–90]. The average number of unique protein identifications from studies using Ion Trap/Orbitrap hybrid instruments as listed in Table 2 is 996 ± 656 (excluding those that used a pre-fractionation step), indicating a marked improvement in proteome coverage as compared to using RP HPLC Ion Trap MS alone.

    • Comprehensive proteome analysis of the response of Pseudomonas putida KT2440 to the flavor compound vanillin

      2014, Journal of Proteomics
      Citation Excerpt :

      For this purpose, we employed a GeLC–MS/MS approach combining protein fractionation by 1-D SDS-PAGE (Supplementary Figure S4) with label free quantification by LC–MS/MS. In total, 1532 proteins with at least two unique peptides and a protein probability of > 99% were identified, corresponding to 29% of the total P. putida proteome. Quantification of these proteins was performed by a label free approach using the fractionation workflow implemented in the Progenesis LCMS software [36]. Proteins that showed a 2-fold or higher change in abundance and a p-value ≤ 0.05 were considered as differentially regulated.

    • Protein expression profiles of chemo-resistant mixed phenotype liver tumors using laser microdissection and LC-MS/MS proteomics

      2013, EuPA Open Proteomics
      Citation Excerpt :

      Due to the characteristic heterogeneity of the tumors and the fact that informative areas can be very focal, the use of frozen tissue samples usually of small size could result in a low yield of representative cases and is therefore not ideal. Large scale global proteomic analysis of laser microdissected FFPE tissue has been successfully employed to discover differentially expressed proteins between different histological tissue types and thus successfully discovered novel protein biomarkers of disease [10–13]. Many of these studies utilized label free quantitative strategies, such as spectral counting and signal intensities of peptide precursor ions, both approaches benefiting from reduced spectral complexity and more analytical depth as well as good linear dynamic range (over two orders of magnitude for spectral counting) and high quantitative proteome coverage [11–15].

    View all citing articles on Scopus
    1

    These authors contributed equally to the work.

    View full text