Recent advances in neuroproteomics and potential application to studies of drug addiction

https://doi.org/10.1016/j.neuropharm.2004.07.009Get rights and content

Abstract

The rapidly growing field of proteomics seeks to track changes in protein expression function that underlie the growth and differentiation of individual cell types, both during normal development and during the onset and progression of disease. Recent years have seen great strides in mRNA expression analysis, and the development of new technologies for protein profiling. However, current methods are limited to analysis of the relative expression level of only a few hundred to perhaps 2000 proteins, well below the ability of DNA microarrays to potentially interrogate the mRNA expression of more than 25 000 genes. Proteomics faces a special challenge in studies of the nervous system, where cellular and sub-cellular architecture is among the most complex in the body. This article presents an overview of current proteomic profiling technologies, reviews the recent use of some of these approaches in studies of the nervous system, and discusses the potential application of neuroproteomics to studies of drug addiction.

Introduction

Recent advances in technology, instrumentation, molecular biology, and bioinformatics have made it possible to begin to analyze entire units of cellular components such as the genome, transcriptome and more recently, the proteome. Although the human genome is estimated to encode for >30 000 proteins, only a fraction of these proteins are expressed in any given cell type. Different proteins vary widely in their level of expression, with some being present in only a few copies while others (e.g., those found in muscles) may be present in millions of copies/cell. Identifying specific cellular proteomes provides opportunities for understanding the physiology of different types of cells and tissues, and as a consequence, how alterations in protein expression are responsible for specific cellular phenotypes. Moreover, identification of cellular proteomes should provide critical information about specific diseases, disease susceptibility and the effects of therapeutic intervention.

The goal of proteomic research is to comprehensively identify all proteins, their associated biological activities, and protein–protein interactions occurring in a given cell. There often is not a direct relationship between the in vivo concentration of an mRNA and its encoded protein (Gygi et al., 1999a, Greenbaum et al., 2003). Differential rates of translation of mRNAs into protein, compartmentalized protein synthesis, and differential rates of protein degradation are factors that confound the extrapolation of mRNA to protein expression profiles. It is estimated that there is an ~100-fold increase in the level of complexity of the proteome compared to the genome that results from differential splicing, protein processing, and post-translational modifications. There is also a huge range in the levels of expression of proteins in different proteomes that limits the ability to analyze all proteins in any given sample. For example, in the serum/plasma proteome there is a 1010 range in plasma protein concentrations (i.e., from 0 to 5 pg/ml for interleukin 6 to 35–50 mg/ml for albumin (Anderson and Anderson, 2002). Unlike mRNA microarray analysis there is no method available to amplify the levels of proteins. Thus, the field of proteomics faces a number of significant technical challenges.

The central nervous system (CNS) poses particular challenges to studying protein function. There is a huge level of cellular heterogeneity, with complex neuronal morphologies and the existence of unique sub-cellular compartments, such as neuronal dendrites, post-synaptic dendritic spines, axons, and pre-synaptic terminals. Alternative splicing is a common feature in the nervous system, and examples include proteins such as neurexin and cadherin, where there is the possibility to generate many thousands of different closely related splice forms from a single gene (Missler and Sudhof, 1998, Wu and Maniatis, 1999). Other examples of alternatively spliced proteins that are particularly important in the CNS include the isoforms of many different neurotransmitter receptors, ion channels, and proteins and enzymes involved in signal transduction. Splicing in neurons may therefore rival the immune system and contribute to the complexity of inter-neuronal communication found in the CNS.

There are many billions of neurons in the CNS, each neuron elaborating often many thousands of synaptic contacts. Thus, intercellular and intracellular signal transduction is a centerpiece of CNS function. It has been estimated that ~20% of human genes are involved in signal transduction (Blume-Jensen and Hunter, 2001). It is not surprising then that many of the close to 600 protein kinases and several 100s of protein phosphatases in the mammalian genome are likely to be expressed in the CNS. As a result, an added complexity of any understanding of neuronal function will require identification of the complex neuronal phosphoproteome.

In this article we briefly review recent advances in proteomic methods and their application to studies of the nervous system. To date, virtually no proteomic approaches have been used in drug addiction research. We therefore briefly discuss several potential applications of neuroproteomic techniques that we believe will be important in future studies of the actions of drugs of abuse.

Section snippets

MS-based protein identification

A common feature of protein profiling methods is their dependence upon proteolytic digestion and analysis by mass spectrometry (MS) for protein identification (Fig. 1). Peptide mass database searching compares experimentally determined peptide masses to theoretical peptide masses calculated from an in silico protein database resulting in a ranked listing of potential proteins in the sample. This approach requires that the masses of a reasonable fraction (e.g., >25%) of the candidate database

Recent studies of neuroproteomics

Analysis of the literature indicates that state-of-the-art proteomic methods have begun in the last 3 years to make some impact in neuroscience (Table 2A–D). The application of these novel techniques will be expected to provide a detailed understanding of biological pathways relevant to the nervous system. Moreover, these studies should accelerate the development of specific diagnostic and prognostic markers of diseases of the nervous system, as well as help in the development of therapeutic

Application of proteomic methods to studies of the actions of drugs of abuse

Gene microarray studies have begun to be used to elucidate some of the transcriptional changes that occur in various animal models of drug abuse (McClung and Nestler, 2003, Sokolov et al., 2003, Yuperov et al., 2003; Yao et al., 2004). However, little is known about the effects of drugs of abuse on the neuronal proteome. Changes in the levels of expression of some “likely suspects” including neurotransmitter receptors, and proteins involved in signal transduction and gene expression, are

Conclusions and future directions

In summary, it is clear from this brief survey that neuroproteomic protein profiling is currently advancing in stride with the emerging, available technologies—with DIGE and ICAT-based protein profiling representing two very attractive and perhaps complementary approaches. In all cases, sample origin and preparation are key concerns for the validity of comparative proteomic analysis. In general, it appears that in terms of sample preparation the more, the better! Prefractionation is key to

References (112)

  • E.R. Marcotte et al.

    cDNA microarray and proteomic approaches in the study of brain diseases: focus on schizophrenia and Alzheimer’s disease

    Pharmacol. Ther

    (2003)
  • D.T. McLachlin et al.

    Analysis of phosphorylated proteins and peptides by mass spectrometry

    Curr. Opin. Chem. Biol

    (2001)
  • K. Mirnics et al.

    Molecular characterization of schizophrenia viewed by microarray analysis of gene expression in prefrontal cortex

    Neuron

    (2000)
  • M. Missler et al.

    Neurexins: three genes and 1001 products

    Trends Genet

    (1998)
  • V.K. Mootha et al.

    Integrated analysis of protein composition, tissue diversity, and gene regulation in mouse mitochondria

    Cell

    (2003)
  • J.V. Olsen et al.

    HysTag—a novel proteomic quantification tool applied to differential display analysis of membrane proteins from distinct areas of mouse brain

    Mol. Cell Proteomics

    (2004)
  • G.J. Opiteck et al.

    Comprehensive two-dimensional high-performance liquid chromatography for the isolation of overexpressed proteins and proteome mapping

    Anal. Biochem

    (1998)
  • J. Peng et al.

    Semiquantitative proteomic analysis of rat forebrain postsynaptic density fractions by mass spectrometry

    J. Biol. Chem

    (2004)
  • E.F. Petricoin et al.

    Use of proteomic patterns in serum to identify ovarian cancer

    Lancet

    (2002)
  • C. Rohlff et al.

    Modern proteomic strategies in the study of complex neuropsychiatric disorders

    Biol. Psychiatr

    (2003)
  • S.M. Stevens et al.

    Proteomic analysis of the synaptic plasma membrane fraction isolated from rat forebrain

    Brain Res. Mol. Brain Res

    (2003)
  • D.B. Wall et al.

    Three-dimensional protein map according to pI, hydrophobicity and molecular mass

    J. Chromatogr. B Anal. Technol. Biomed. Life Sci

    (2002)
  • Q. Wu et al.

    A striking organization of a large family of human neural cadherin_like cell adhesion genes

    Cell

    (1999)
  • B.L. Adam et al.

    Serum protein fingerprinting coupled with a pattern-matching algorithm distinguishes prostate cancer from benign prostate hyperplasia and healthy men

    Cancer Res

    (2002)
  • R. Aebersold

    Constellations in a cellular universe

    Nature

    (2003)
  • C. Becamel et al.

    Synaptic multiprotein complexes associated with 5-HT2C receptors: a proteomic approach

    EMBO J

    (2002)
  • E. Betgovargez et al.

    Reproducibility and dynamic range characteristics of the proteome PF 2D system

    Beckman Coulter Application Information Bulletin A-1964A

    (2003)
  • Blume-Jensen, P., Hunter, T., 2001. Oncogenic kinase signalling. Nature 411,...
  • J.A. Bibb et al.

    Effects of chronic exposure to cocaine are regulated by the neuronal protein Cdk5

    Nature

    (2001)
  • M.S. Boguski et al.

    Biomedical informatics for proteomics

    Nature

    (2003)
  • D. Bonenfant et al.

    Quantitation of changes in protein phosphorylation: a simple method based on stable isotope labeling and mass spectrometry

    Proc. Natl. Acad. Sci. USA

    (2003)
  • M.M. Bushey et al.

    Automated instrumentation for comprehensive two-dimensional high-performance liquid chromatography of proteins

    Anal. Chem

    (1990)
  • O. Carrette et al.

    A panel of cerebrospinal fluid potential biomarkers for the diagnosis of Alzheimer’s disease

    Proteomics

    (2003)
  • B.E. Chong et al.

    Chromatofocusing nonporous reversed-phase high-performance liquid chromatography/electrospray ionization time-of-flight mass spectrometry of proteins from human breast cancer whole cell lysates

    Rapid Commun. Mass Spectrom

    (2001)
  • J. Choudhary et al.

    Proteomics in postgenomic neuroscience: the end of the beginning

    Nat. Neurosci

    (2004)
  • P. Cohen

    The role of protein phosphorylation in neural and hormonal control of cellular activity

    Nature

    (1982)
  • S. Ficarro et al.

    Phosphproteome analysis by mass spectrometry and its application to Saccharomyces cerevisiae

    Nat. Biotechnol

    (2002)
  • J. Flora et al.

    Selective, sensitive, and rapid phosphopeptide identification in enzymatic digests using ESI-FTICR-MS with infrared multiphoton dissociation

    Anal. Chem

    (2001)
  • M. Fountoulakis et al.

    Proteomic analysis of the fetal brain

    Proteomics

    (2002)
  • W.M. Freeman et al.

    Proteomics for protein expression profiling in neuroscience

    Neurochem. Res

    (2004)
  • M.B. Goshe et al.

    Phosphoprotein isotope-coded affinity tag approach for isolating and quantitating phosphopeptides in proteome-wide analyses

    Anal. Chem

    (2001)
  • M.B. Goshe et al.

    Phosphoprotein isotope-coded affinity tags: application to the enrichment and identification of low-abundance phosphoproteins

    Anal. Chem

    (2002)
  • D. Greenbaum et al.

    Comparing protein abundance and mRNA expression levels on a genomic scale

    Genome Biol

    (2003)
  • S.P. Gygi et al.

    Correlation between protein and mRNA abundance in yeast

    Mol. Cell Biol

    (1999)
  • S.P. Gygi et al.

    Quantitative analysis of complex protein mixtures using isotope-coded affinity tags

    Nat. Biotechnol

    (1999)
  • S.P. Gygi et al.

    Evaluation of two-dimensional gel electrophoresis-based proteome analysis technology

    Proc. Natl. Acad. Sci. USA

    (2000)
  • B.B. Haab et al.

    Protein microarrays for highly parallel detection and quantitation of specific proteins and antibodies in complex solutions

    Genome Biol

    (2001)
  • K. Hakansson et al.

    Electron capture dissociation and infrared multiphoton dissociation MS/MS of an N-glycosylated tryptic peptide to yield complementary sequence information

    Anal. Chem

    (2001)
  • D.K. Han et al.

    Quantitative profiling of differentiation-induced microsomal proteins using isotope-coded affinity tags and mass spectrometry

    Nat. Biotechnol

    (2001)
  • H. Husi et al.

    Proteomic analysis of NMDA receptor-adhesion protein signaling complexes

    Nat. Neurosci

    (2000)
  • Cited by (0)

    View full text