Immunocytochemistry

Immunocytochemistry (ICC) is a technique for detection and visualization of proteins, or other antigens, in cells using antibodies specifically recognizing the target of interest. The antibody is directly or indirectly linked to a reporter, such as a fluorophore or enzyme. The reporter gives rise to a signal, such as fluorescence or color from an enzymatic reaction, which can be detected in a microscope. The type of microscope used depends on the type of reporter. In ICC, the staining technique is applied on cultured cells or individual cells that have been isolated from eg. tissues, blood samples or mouth swabs. This is in contrast to immunohistochemistry (IHC), where cells are analyzed within intact tissue sections.

Technology

Immunocytochemistry is usually performed in four sequential steps. First, the cells are seeded on a solid support, which is usually a glass slide or a glass-bottom plate. Depending on the type of cells and seeding technique, an incubation time might be necessary before proceeding with immunostaining. In case of seeding adherent cells, the cells will attach to the solid support surface during the incubation, which varies from half an hour to 24 h for the different cell types. In the second step, the cells are subjected to immunostaining, which involves fixation, permeabilization, and antibody incubation. Fixation retains the proteins at their location in the cell and preserves their chemical and structural state at the time of fixation. It can be done by crosslinking or by precipitating the proteins using organic solvents. Upon permeabilization, membranes are punctured with the use of solvents or detergents, allowing the relatively large antibodies to cross the cellular membranes. The permeabilization requires fixation, and hence limits the technique to studying dead cells. During antibody incubation, the antibodies are allowed to bind to target antigens within the cells, after which unbound antibodies are removed by washing. In the third step, the cells and the locations of antibodies bound to target antigens are visualized using microscopy. Images are acquired using a camera or other detector, and in the final step, the images are analyzed and cellular structures annotated. Figure 1 describes a typical workflow for ICC using a fluorescent reporter.


Figure 1. The four steps of immunocytochemistry: (i) cell seeding, (ii) immunostaining, (iii) imaging, and (iv) image analysis.

Reporters

As for IHC, there are different reporter systems available for ICC. One is the use of enzyme-coupled antibodies. After the addition of a substrate, the enzyme catalyzes a reaction that generates a coloured product at the site where the enzyme-coupled antibody is bound in the cells. For example, the commonly used enzyme horseradish peroxidase (HRP) can convert 3,3'-diaminobenzidine (DAB) into a brown precipitate, which can be detected using light-microscopy. Another type of reporter is fluorophores. These molecules can be transiently excited to a higher energy state upon absorption of light with a particular wavelength, and thereafter relax to the ground state while emitting light of a longer wavelength. In this case, a fluorescence microscope is used to excite the fluorophores as well as detecting their emission. Since different fluorophores are excited by different wavelengths of light and also emit light at different wavelengths, multiple fluorophores with different colors may be combined in the same sample. This enables the acquisition of multicolor images, where each color represents a specific antigen target. However, the number of fluorophores used in the same sample is limited by the spectral overlap of the excitation and emission profiles of the fluorophores, as the signals from fluorophores with similar spectral properties cannot readily be separated. In addition to fluorophore-labeled antibodies, there are molecules that are fluorescent by themselves and have intrinsic ability to bind specifically to other molecules. These molecules may be used together with the fluorophore-labeled antibodies. One example is 4',6-diamidino-2-phenylindole (DAPI), which binds to DNA and is commonly used to visualize the cell nucleus. DAPI is excited by ultraviolet light and then emits light in the blue spectrum. A consideration when using fluorophores as reporters is that bleaching will occur when the fluorophores are exposed to light. Over time, the stained sample will decrease in brightness.

Table 1. Examples of different reporters

Reporter type Reporter example Visualization Specificity
Enzyme-coupled antibody Antibody-peroxidase + DAB Brown color Antigen
Fluorophore-labeled antibody Antibody-Cy3 Green excitation / yellow emission Antigen
Biospecific small molecule dye DAPI UV excitation / blue emission DNA

Direct vs. indirect detection

The detection method for the immunostaining can be either direct or indirect. In the direct method, the molecule of interest is directly targeted by a primary antibody linked to the reporter, giving a rapid and specific method. However, it is usually not sensitive enough for most proteins as the number of present copies of the protein is too low to yield a strong enough signal. In the indirect method, the molecule of interest is targeted by an un-labelled primary antibody, which is in turn detected using a reporter-coupled secondary antibody that recognizes the primary antibody (see Figure 1ii). The indirect method is more sensitive due to binding of multiple secondary antibodies to each primary antibody, resulting in signal amplification. Another advantage is also an increased flexibility because of the possibility to vary the primary and secondary antibody combination. Also, since the secondary antibody is targeting the constant region of the primary antibody, which is species-specific, the same secondary antibody can be used for all primary antibodies raised in a given species. The disadvantages of the indirect method are a more laborious and time-consuming protocol, and a risk of non-specific binding of the secondary antibody.

Specific examples

In the Human Protein Atlas, ICC with fluorescence as a reporter (ICC-IF) is used to analyze the subcellular distribution of proteins and build a Cell Atlas of the whole human proteome (Barbe L et al. (2008)). For each protein the subcellular localization is studied in three different human cell lines, mainly using antibodies produced within the Human Protein Atlas project. Cells cultured in vitro, are fixed with paraformaldehyde, permeabilized by treatment with the detergent Triton X-100, and stained by indirect immunofluorescence (Stadler C et al. (2010)). In addition to the antibody targeting the protein of interest, two reference marker antibodies are used to stain the endoplasmic reticulum and microtubules, respectively. and the cells are also counterstained with the nuclear probe DAPI. A confocal laser scanning microscope equipped with a 63x magnification oil immersion objective is used to acquire high-resolution images of the stainings. The images are manually annotated to provide a description of subcellular localization, staining characteristics, and staining intensity. Furthermore, each location is given a reliability score in order to indicate if the results are supported by external experimental data or internal antibody validation. In the end, a knowledge-based revision of the subcellular distribution is performed in a gene-centric manner, taking into account the staining of one or multiple antibodies. Figure 2 shows typical results from ICC-IF in the Cell Atlas.

Figure 2a. RNA binding motif protein 25 (RBM25) localized to nuclear speckles (green). Microtubules are stained in red.


Figure 2b. Golgin B1 (GOLGB1) localized to the Golgi apparatus (green). Microtubules are stained in red and the nucleus in blue (DAPI).


Figure 2c. Electron-transfer-flavoprotein, alpha polypeptide (ETFA) localized to mitochondria (green). Microtubules are stained in red, nucleus in blue (DAPI).


In addition to ICC-IFs, the Human Protein Atlas project has also analyzed protein expression patterns in a panel of cell lines using cell microarrays (Andersson et al., 2006). For preparation of the microarrays, the cells of the different cell lines were fixed in formaldehyde, dispersed in agarose, embedded in paraffin and placed on glass slides. Analysis of protein expression was carried out using Human Protein Atlas generated primary antibodies, HRP-coupled secondary antibodies, and DAB substrate, gresulting in a brown precipitate that correlates with the protein expression. The cells were also counterstained with hematoxylin to give a general staining of the cell structure.

References and Links

Parikh K et al., Colonic epithelial cell diversity in health and inflammatory bowel disease. Nature. (2019)
PubMed: 30814735 DOI: 10.1038/s41586-019-0992-y

Menon M et al., Single-cell transcriptomic atlas of the human retina identifies cell types associated with age-related macular degeneration. Nat Commun. (2019)
PubMed: 31653841 DOI: 10.1038/s41467-019-12780-8

Wang L et al., Single-cell reconstruction of the adult human heart during heart failure and recovery reveals the cellular landscape underlying cardiac function. Nat Cell Biol. (2020)
PubMed: 31915373 DOI: 10.1038/s41556-019-0446-7

Wang Y et al., Single-cell transcriptome analysis reveals differential nutrient absorption functions in human intestine. J Exp Med. (2020)
PubMed: 31753849 DOI: 10.1084/jem.20191130

Liao J et al., Single-cell RNA sequencing of human kidney. Sci Data. (2020)
PubMed: 31896769 DOI: 10.1038/s41597-019-0351-8

MacParland SA et al., Single cell RNA sequencing of human liver reveals distinct intrahepatic macrophage populations. Nat Commun. (2018)
PubMed: 30348985 DOI: 10.1038/s41467-018-06318-7

Vieira Braga FA et al., A cellular census of human lungs identifies novel cell states in health and in asthma. Nat Med. (2019)
PubMed: 31209336 DOI: 10.1038/s41591-019-0468-5

Vento-Tormo R et al., Single-cell reconstruction of the early maternal-fetal interface in humans. Nature. (2018)
PubMed: 30429548 DOI: 10.1038/s41586-018-0698-6

Qadir MMF et al., Single-cell resolution analysis of the human pancreatic ductal progenitor cell niche. Proc Natl Acad Sci U S A. (2020)
PubMed: 32354994 DOI: 10.1073/pnas.1918314117

Solé-Boldo L et al., Single-cell transcriptomes of the human skin reveal age-related loss of fibroblast priming. Commun Biol. (2020)
PubMed: 32327715 DOI: 10.1038/s42003-020-0922-4

Henry GH et al., A Cellular Anatomy of the Normal Adult Human Prostate and Prostatic Urethra. Cell Rep. (2018)
PubMed: 30566875 DOI: 10.1016/j.celrep.2018.11.086

Chen J et al., PBMC fixation and processing for Chromium single-cell RNA sequencing. J Transl Med. (2018)
PubMed: 30016977 DOI: 10.1186/s12967-018-1578-4

Guo J et al., The adult human testis transcriptional cell atlas. Cell Res. (2018)
PubMed: 30315278 DOI: 10.1038/s41422-018-0099-2

Uhlen M et al., A proposal for validation of antibodies. Nat Methods. (2016)
PubMed: 27595404 DOI: 10.1038/nmeth.3995

Stadler C et al., Systematic validation of antibody binding and protein subcellular localization using siRNA and confocal microscopy. J Proteomics. (2012)
PubMed: 22361696 DOI: 10.1016/j.jprot.2012.01.030

Poser I et al., BAC TransgeneOmics: a high-throughput method for exploration of protein function in mammals. Nat Methods. (2008)
PubMed: 18391959 DOI: 10.1038/nmeth.1199

Skogs M et al., Antibody Validation in Bioimaging Applications Based on Endogenous Expression of Tagged Proteins. J Proteome Res. (2017)
PubMed: 27723985 DOI: 10.1021/acs.jproteome.6b00821

Takahashi H et al., 5' end-centered expression profiling using cap-analysis gene expression and next-generation sequencing. Nat Protoc. (2012)
PubMed: 22362160 DOI: 10.1038/nprot.2012.005

Lein ES et al., Genome-wide atlas of gene expression in the adult mouse brain. Nature. (2007)
PubMed: 17151600 DOI: 10.1038/nature05453

Kircher M et al., Double indexing overcomes inaccuracies in multiplex sequencing on the Illumina platform. Nucleic Acids Res. (2012)
PubMed: 22021376 DOI: 10.1093/nar/gkr771

Pollard TD et al., Actin, a central player in cell shape and movement. Science. (2009)
PubMed: 19965462 DOI: 10.1126/science.1175862

Mitchison TJ et al., Actin-based cell motility and cell locomotion. Cell. (1996)
PubMed: 8608590 

Pollard TD et al., Molecular Mechanism of Cytokinesis. Annu Rev Biochem. (2019)
PubMed: 30649923 DOI: 10.1146/annurev-biochem-062917-012530

dos Remedios CG et al., Actin binding proteins: regulation of cytoskeletal microfilaments. Physiol Rev. (2003)
PubMed: 12663865 DOI: 10.1152/physrev.00026.2002

Campellone KG et al., A nucleator arms race: cellular control of actin assembly. Nat Rev Mol Cell Biol. (2010)
PubMed: 20237478 DOI: 10.1038/nrm2867

Rottner K et al., Actin assembly mechanisms at a glance. J Cell Sci. (2017)
PubMed: 29032357 DOI: 10.1242/jcs.206433

Bird RP., Observation and quantification of aberrant crypts in the murine colon treated with a colon carcinogen: preliminary findings. Cancer Lett. (1987)
PubMed: 3677050 DOI: 10.1016/0304-3835(87)90157-1

HUXLEY AF et al., Structural changes in muscle during contraction; interference microscopy of living muscle fibres. Nature. (1954)
PubMed: 13165697 

HUXLEY H et al., Changes in the cross-striations of muscle during contraction and stretch and their structural interpretation. Nature. (1954)
PubMed: 13165698 

Svitkina T., The Actin Cytoskeleton and Actin-Based Motility. Cold Spring Harb Perspect Biol. (2018)
PubMed: 29295889 DOI: 10.1101/cshperspect.a018267

Kelpsch DJ et al., Nuclear Actin: From Discovery to Function. Anat Rec (Hoboken). (2018)
PubMed: 30312531 DOI: 10.1002/ar.23959

Malumbres M et al., Cell cycle, CDKs and cancer: a changing paradigm. Nat Rev Cancer. (2009)
PubMed: 19238148 DOI: 10.1038/nrc2602

Massagué J., G1 cell-cycle control and cancer. Nature. (2004)
PubMed: 15549091 DOI: 10.1038/nature03094

Hartwell LH et al., Cell cycle control and cancer. Science. (1994)
PubMed: 7997877 DOI: 10.1126/science.7997877

Barnum KJ et al., Cell cycle regulation by checkpoints. Methods Mol Biol. (2014)
PubMed: 24906307 DOI: 10.1007/978-1-4939-0888-2_2

Weinberg RA., The retinoblastoma protein and cell cycle control. Cell. (1995)
PubMed: 7736585 DOI: 10.1016/0092-8674(95)90385-2

Morgan DO., Principles of CDK regulation. Nature. (1995)
PubMed: 7877684 DOI: 10.1038/374131a0

Teixeira LK et al., Ubiquitin ligases and cell cycle control. Annu Rev Biochem. (2013)
PubMed: 23495935 DOI: 10.1146/annurev-biochem-060410-105307

King RW et al., How proteolysis drives the cell cycle. Science. (1996)
PubMed: 8939846 DOI: 10.1126/science.274.5293.1652

Cho RJ et al., Transcriptional regulation and function during the human cell cycle. Nat Genet. (2001)
PubMed: 11137997 DOI: 10.1038/83751

Whitfield ML et al., Identification of genes periodically expressed in the human cell cycle and their expression in tumors. Mol Biol Cell. (2002)
PubMed: 12058064 DOI: 10.1091/mbc.02-02-0030.

Boström J et al., Comparative cell cycle transcriptomics reveals synchronization of developmental transcription factor networks in cancer cells. PLoS One. (2017)
PubMed: 29228002 DOI: 10.1371/journal.pone.0188772

Lane KR et al., Cell cycle-regulated protein abundance changes in synchronously proliferating HeLa cells include regulation of pre-mRNA splicing proteins. PLoS One. (2013)
PubMed: 23520512 DOI: 10.1371/journal.pone.0058456

Ohta S et al., The protein composition of mitotic chromosomes determined using multiclassifier combinatorial proteomics. Cell. (2010)
PubMed: 20813266 DOI: 10.1016/j.cell.2010.07.047

Ly T et al., A proteomic chronology of gene expression through the cell cycle in human myeloid leukemia cells. Elife. (2014)
PubMed: 24596151 DOI: 10.7554/eLife.01630

Pagliuca FW et al., Quantitative proteomics reveals the basis for the biochemical specificity of the cell-cycle machinery. Mol Cell. (2011)
PubMed: 21816347 DOI: 10.1016/j.molcel.2011.05.031

Ly T et al., Proteomic analysis of the response to cell cycle arrests in human myeloid leukemia cells. Elife. (2015)
PubMed: 25555159 DOI: 10.7554/eLife.04534

Dueck H et al., Variation is function: Are single cell differences functionally important?: Testing the hypothesis that single cell variation is required for aggregate function. Bioessays. (2016)
PubMed: 26625861 DOI: 10.1002/bies.201500124

Snijder B et al., Origins of regulated cell-to-cell variability. Nat Rev Mol Cell Biol. (2011)
PubMed: 21224886 DOI: 10.1038/nrm3044

Thul PJ et al., A subcellular map of the human proteome. Science. (2017)
PubMed: 28495876 DOI: 10.1126/science.aal3321

Cooper S et al., Membrane-elution analysis of content of cyclins A, B1, and E during the unperturbed mammalian cell cycle. Cell Div. (2007)
PubMed: 17892542 DOI: 10.1186/1747-1028-2-28

Davis PK et al., Biological methods for cell-cycle synchronization of mammalian cells. Biotechniques. (2001)
PubMed: 11414226 DOI: 10.2144/01306rv01

Domenighetti G et al., Effect of information campaign by the mass media on hysterectomy rates. Lancet. (1988)
PubMed: 2904581 DOI: 10.1016/s0140-6736(88)90943-9

Scialdone A et al., Computational assignment of cell-cycle stage from single-cell transcriptome data. Methods. (2015)
PubMed: 26142758 DOI: 10.1016/j.ymeth.2015.06.021

Sakaue-Sawano A et al., Visualizing spatiotemporal dynamics of multicellular cell-cycle progression. Cell. (2008)
PubMed: 18267078 DOI: 10.1016/j.cell.2007.12.033

Grant GD et al., Identification of cell cycle-regulated genes periodically expressed in U2OS cells and their regulation by FOXM1 and E2F transcription factors. Mol Biol Cell. (2013)
PubMed: 24109597 DOI: 10.1091/mbc.E13-05-0264

Semple JW et al., An essential role for Orc6 in DNA replication through maintenance of pre-replicative complexes. EMBO J. (2006)
PubMed: 17053779 DOI: 10.1038/sj.emboj.7601391

Kilfoil ML et al., Stochastic variation: from single cells to superorganisms. HFSP J. (2009)
PubMed: 20514130 DOI: 10.2976/1.3223356

Ansel J et al., Cell-to-cell stochastic variation in gene expression is a complex genetic trait. PLoS Genet. (2008)
PubMed: 18404214 DOI: 10.1371/journal.pgen.1000049

Colman-Lerner A et al., Regulated cell-to-cell variation in a cell-fate decision system. Nature. (2005)
PubMed: 16170311 DOI: 10.1038/nature03998

Liberali P et al., Single-cell and multivariate approaches in genetic perturbation screens. Nat Rev Genet. (2015)
PubMed: 25446316 DOI: 10.1038/nrg3768

Elowitz MB et al., Stochastic gene expression in a single cell. Science. (2002)
PubMed: 12183631 DOI: 10.1126/science.1070919

Kaern M et al., Stochasticity in gene expression: from theories to phenotypes. Nat Rev Genet. (2005)
PubMed: 15883588 DOI: 10.1038/nrg1615

Bianconi E et al., An estimation of the number of cells in the human body. Ann Hum Biol. (2013)
PubMed: 23829164 DOI: 10.3109/03014460.2013.807878

Malumbres M., Cyclin-dependent kinases. Genome Biol. (2014)
PubMed: 25180339 

Collins K et al., The cell cycle and cancer. Proc Natl Acad Sci U S A. (1997)
PubMed: 9096291 

Zhivotovsky B et al., Cell cycle and cell death in disease: past, present and future. J Intern Med. (2010)
PubMed: 20964732 DOI: 10.1111/j.1365-2796.2010.02282.x

Cho RJ et al., A genome-wide transcriptional analysis of the mitotic cell cycle. Mol Cell. (1998)
PubMed: 9702192 

Spellman PT et al., Comprehensive identification of cell cycle-regulated genes of the yeast Saccharomyces cerevisiae by microarray hybridization. Mol Biol Cell. (1998)
PubMed: 9843569 

Orlando DA et al., Global control of cell-cycle transcription by coupled CDK and network oscillators. Nature. (2008)
PubMed: 18463633 DOI: 10.1038/nature06955

Rustici G et al., Periodic gene expression program of the fission yeast cell cycle. Nat Genet. (2004)
PubMed: 15195092 DOI: 10.1038/ng1377

Uhlén M et al., Tissue-based map of the human proteome. Science (2015)
PubMed: 25613900 DOI: 10.1126/science.1260419

Nigg EA et al., The centrosome cycle: Centriole biogenesis, duplication and inherent asymmetries. Nat Cell Biol. (2011)
PubMed: 21968988 DOI: 10.1038/ncb2345

Doxsey S., Re-evaluating centrosome function. Nat Rev Mol Cell Biol. (2001)
PubMed: 11533726 DOI: 10.1038/35089575

Bornens M., Centrosome composition and microtubule anchoring mechanisms. Curr Opin Cell Biol. (2002)
PubMed: 11792541 

Conduit PT et al., Centrosome function and assembly in animal cells. Nat Rev Mol Cell Biol. (2015)
PubMed: 26373263 DOI: 10.1038/nrm4062

Tollenaere MA et al., Centriolar satellites: key mediators of centrosome functions. Cell Mol Life Sci. (2015)
PubMed: 25173771 DOI: 10.1007/s00018-014-1711-3

Prosser SL et al., Centriolar satellite biogenesis and function in vertebrate cells. J Cell Sci. (2020)
PubMed: 31896603 DOI: 10.1242/jcs.239566

Rieder CL et al., The centrosome in vertebrates: more than a microtubule-organizing center. Trends Cell Biol. (2001)
PubMed: 11567874 

Badano JL et al., The centrosome in human genetic disease. Nat Rev Genet. (2005)
PubMed: 15738963 DOI: 10.1038/nrg1557

Clegg JS., Properties and metabolism of the aqueous cytoplasm and its boundaries. Am J Physiol. (1984)
PubMed: 6364846 

Luby-Phelps K., The physical chemistry of cytoplasm and its influence on cell function: an update. Mol Biol Cell. (2013)
PubMed: 23989722 DOI: 10.1091/mbc.E12-08-0617

Luby-Phelps K., Cytoarchitecture and physical properties of cytoplasm: volume, viscosity, diffusion, intracellular surface area. Int Rev Cytol. (2000)
PubMed: 10553280 

Ellis RJ., Macromolecular crowding: obvious but underappreciated. Trends Biochem Sci. (2001)
PubMed: 11590012 

Bright GR et al., Fluorescence ratio imaging microscopy: temporal and spatial measurements of cytoplasmic pH. J Cell Biol. (1987)
PubMed: 3558476 

Kopito RR., Aggresomes, inclusion bodies and protein aggregation. Trends Cell Biol. (2000)
PubMed: 11121744 

Aizer A et al., Intracellular trafficking and dynamics of P bodies. Prion. (2008)
PubMed: 19242093 

Carcamo WC et al., Molecular cell biology and immunobiology of mammalian rod/ring structures. Int Rev Cell Mol Biol. (2014)
PubMed: 24411169 DOI: 10.1016/B978-0-12-800097-7.00002-6

Lang F., Mechanisms and significance of cell volume regulation. J Am Coll Nutr. (2007)
PubMed: 17921474 

Schwarz DS et al., The endoplasmic reticulum: structure, function and response to cellular signaling. Cell Mol Life Sci. (2016)
PubMed: 26433683 DOI: 10.1007/s00018-015-2052-6

Friedman JR et al., The ER in 3D: a multifunctional dynamic membrane network. Trends Cell Biol. (2011)
PubMed: 21900009 DOI: 10.1016/j.tcb.2011.07.004

Travers KJ et al., Functional and genomic analyses reveal an essential coordination between the unfolded protein response and ER-associated degradation. Cell. (2000)
PubMed: 10847680 

Roussel BD et al., Endoplasmic reticulum dysfunction in neurological disease. Lancet Neurol. (2013)
PubMed: 23237905 DOI: 10.1016/S1474-4422(12)70238-7

Neve EP et al., Cytochrome P450 proteins: retention and distribution from the endoplasmic reticulum. Curr Opin Drug Discov Devel. (2010)
PubMed: 20047148 

Kulkarni-Gosavi P et al., Form and function of the Golgi apparatus: scaffolds, cytoskeleton and signalling. FEBS Lett. (2019)
PubMed: 31378930 DOI: 10.1002/1873-3468.13567

Short B et al., The Golgi apparatus. Curr Biol. (2000)
PubMed: 10985372 DOI: 10.1016/s0960-9822(00)00644-8

Wei JH et al., Unraveling the Golgi ribbon. Traffic. (2010)
PubMed: 21040294 DOI: 10.1111/j.1600-0854.2010.01114.x

Wilson C et al., The Golgi apparatus: an organelle with multiple complex functions. Biochem J. (2011)
PubMed: 21158737 DOI: 10.1042/BJ20101058

Farquhar MG et al., The Golgi apparatus: 100 years of progress and controversy. Trends Cell Biol. (1998)
PubMed: 9695800 

Brandizzi F et al., Organization of the ER-Golgi interface for membrane traffic control. Nat Rev Mol Cell Biol. (2013)
PubMed: 23698585 DOI: 10.1038/nrm3588

Potelle S et al., Golgi post-translational modifications and associated diseases. J Inherit Metab Dis. (2015)
PubMed: 25967285 DOI: 10.1007/s10545-015-9851-7

Leduc C et al., Intermediate filaments in cell migration and invasion: the unusual suspects. Curr Opin Cell Biol. (2015)
PubMed: 25660489 DOI: 10.1016/j.ceb.2015.01.005

Lowery J et al., Intermediate Filaments Play a Pivotal Role in Regulating Cell Architecture and Function. J Biol Chem. (2015)
PubMed: 25957409 DOI: 10.1074/jbc.R115.640359

Robert A et al., Intermediate filament dynamics: What we can see now and why it matters. Bioessays. (2016)
PubMed: 26763143 DOI: 10.1002/bies.201500142

Fuchs E et al., Intermediate filaments: structure, dynamics, function, and disease. Annu Rev Biochem. (1994)
PubMed: 7979242 DOI: 10.1146/annurev.bi.63.070194.002021

Janmey PA et al., Viscoelastic properties of vimentin compared with other filamentous biopolymer networks. J Cell Biol. (1991)
PubMed: 2007620 

Köster S et al., Intermediate filament mechanics in vitro and in the cell: from coiled coils to filaments, fibers and networks. Curr Opin Cell Biol. (2015)
PubMed: 25621895 DOI: 10.1016/j.ceb.2015.01.001

Herrmann H et al., Intermediate filaments: from cell architecture to nanomechanics. Nat Rev Mol Cell Biol. (2007)
PubMed: 17551517 DOI: 10.1038/nrm2197

Gauster M et al., Keratins in the human trophoblast. Histol Histopathol. (2013)
PubMed: 23450430 DOI: 10.14670/HH-28.817

Janke C., The tubulin code: molecular components, readout mechanisms, and functions. J Cell Biol. (2014)
PubMed: 25135932 DOI: 10.1083/jcb.201406055

Goodson HV et al., Microtubules and Microtubule-Associated Proteins. Cold Spring Harb Perspect Biol. (2018)
PubMed: 29858272 DOI: 10.1101/cshperspect.a022608

Wade RH., On and around microtubules: an overview. Mol Biotechnol. (2009)
PubMed: 19565362 DOI: 10.1007/s12033-009-9193-5

Desai A et al., Microtubule polymerization dynamics. Annu Rev Cell Dev Biol. (1997)
PubMed: 9442869 DOI: 10.1146/annurev.cellbio.13.1.83

Conde C et al., Microtubule assembly, organization and dynamics in axons and dendrites. Nat Rev Neurosci. (2009)
PubMed: 19377501 DOI: 10.1038/nrn2631

Wloga D et al., Post-translational modifications of microtubules. J Cell Sci. (2010)
PubMed: 20930140 DOI: 10.1242/jcs.063727

Schmoranzer J et al., Role of microtubules in fusion of post-Golgi vesicles to the plasma membrane. Mol Biol Cell. (2003)
PubMed: 12686609 DOI: 10.1091/mbc.E02-08-0500

Skop AR et al., Dissection of the mammalian midbody proteome reveals conserved cytokinesis mechanisms. Science. (2004)
PubMed: 15166316 DOI: 10.1126/science.1097931

Waters AM et al., Ciliopathies: an expanding disease spectrum. Pediatr Nephrol. (2011)
PubMed: 21210154 DOI: 10.1007/s00467-010-1731-7

Matamoros AJ et al., Microtubules in health and degenerative disease of the nervous system. Brain Res Bull. (2016)
PubMed: 27365230 DOI: 10.1016/j.brainresbull.2016.06.016

Jordan MA et al., Microtubules as a target for anticancer drugs. Nat Rev Cancer. (2004)
PubMed: 15057285 DOI: 10.1038/nrc1317

Nunnari J et al., Mitochondria: in sickness and in health. Cell. (2012)
PubMed: 22424226 DOI: 10.1016/j.cell.2012.02.035

Friedman JR et al., Mitochondrial form and function. Nature. (2014)
PubMed: 24429632 DOI: 10.1038/nature12985

Calvo SE et al., The mitochondrial proteome and human disease. Annu Rev Genomics Hum Genet. (2010)
PubMed: 20690818 DOI: 10.1146/annurev-genom-082509-141720

McBride HM et al., Mitochondria: more than just a powerhouse. Curr Biol. (2006)
PubMed: 16860735 DOI: 10.1016/j.cub.2006.06.054

Schaefer AM et al., The epidemiology of mitochondrial disorders--past, present and future. Biochim Biophys Acta. (2004)
PubMed: 15576042 DOI: 10.1016/j.bbabio.2004.09.005

Lange A et al., Classical nuclear localization signals: definition, function, and interaction with importin alpha. J Biol Chem. (2007)
PubMed: 17170104 DOI: 10.1074/jbc.R600026200

Ashmarina LI et al., 3-Hydroxy-3-methylglutaryl coenzyme A lyase: targeting and processing in peroxisomes and mitochondria. J Lipid Res. (1999)
PubMed: 9869651 

Wang SC et al., Nuclear translocation of the epidermal growth factor receptor family membrane tyrosine kinase receptors. Clin Cancer Res. (2009)
PubMed: 19861462 DOI: 10.1158/1078-0432.CCR-08-2813

Jeffery CJ., Moonlighting proteins. Trends Biochem Sci. (1999)
PubMed: 10087914 

Jeffery CJ., Why study moonlighting proteins? Front Genet. (2015)
PubMed: 26150826 DOI: 10.3389/fgene.2015.00211

Pancholi V., Multifunctional alpha-enolase: its role in diseases. Cell Mol Life Sci. (2001)
PubMed: 11497239 DOI: 10.1007/pl00000910

Chapple CE et al., Extreme multifunctional proteins identified from a human protein interaction network. Nat Commun. (2015)
PubMed: 26054620 DOI: 10.1038/ncomms8412

Dechat T et al., Nuclear lamins: major factors in the structural organization and function of the nucleus and chromatin. Genes Dev. (2008)
PubMed: 18381888 DOI: 10.1101/gad.1652708

Gruenbaum Y et al., The nuclear lamina comes of age. Nat Rev Mol Cell Biol. (2005)
PubMed: 15688064 DOI: 10.1038/nrm1550

Stuurman N et al., Nuclear lamins: their structure, assembly, and interactions. J Struct Biol. (1998)
PubMed: 9724605 DOI: 10.1006/jsbi.1998.3987

Paine PL et al., Nuclear envelope permeability. Nature. (1975)
PubMed: 1117994 

Reichelt R et al., Correlation between structure and mass distribution of the nuclear pore complex and of distinct pore complex components. J Cell Biol. (1990)
PubMed: 2324201 

CALLAN HG et al., Experimental studies on amphibian oocyte nuclei. I. Investigation of the structure of the nuclear membrane by means of the electron microscope. Proc R Soc Lond B Biol Sci. (1950)
PubMed: 14786306 

WATSON ML., The nuclear envelope; its structure and relation to cytoplasmic membranes. J Biophys Biochem Cytol. (1955)
PubMed: 13242591 

BAHR GF et al., The fine structure of the nuclear membrane in the larval salivary gland and midgut of Chironomus. Exp Cell Res. (1954)
PubMed: 13173504 

Terasaki M et al., A new model for nuclear envelope breakdown. Mol Biol Cell. (2001)
PubMed: 11179431 

Dultz E et al., Systematic kinetic analysis of mitotic dis- and reassembly of the nuclear pore in living cells. J Cell Biol. (2008)
PubMed: 18316408 DOI: 10.1083/jcb.200707026

Salina D et al., Cytoplasmic dynein as a facilitator of nuclear envelope breakdown. Cell. (2002)
PubMed: 11792324 

Beaudouin J et al., Nuclear envelope breakdown proceeds by microtubule-induced tearing of the lamina. Cell. (2002)
PubMed: 11792323 

Gerace L et al., The nuclear envelope lamina is reversibly depolymerized during mitosis. Cell. (1980)
PubMed: 7357605 

Ellenberg J et al., Nuclear membrane dynamics and reassembly in living cells: targeting of an inner nuclear membrane protein in interphase and mitosis. J Cell Biol. (1997)
PubMed: 9298976 

Yang L et al., Integral membrane proteins of the nuclear envelope are dispersed throughout the endoplasmic reticulum during mitosis. J Cell Biol. (1997)
PubMed: 9182656 

Bione S et al., Identification of a novel X-linked gene responsible for Emery-Dreifuss muscular dystrophy. Nat Genet. (1994)
PubMed: 7894480 DOI: 10.1038/ng1294-323

Boisvert FM et al., The multifunctional nucleolus. Nat Rev Mol Cell Biol. (2007)
PubMed: 17519961 DOI: 10.1038/nrm2184

Scheer U et al., Structure and function of the nucleolus. Curr Opin Cell Biol. (1999)
PubMed: 10395554 DOI: 10.1016/S0955-0674(99)80054-4

Németh A et al., Genome organization in and around the nucleolus. Trends Genet. (2011)
PubMed: 21295884 DOI: 10.1016/j.tig.2011.01.002

Cuylen S et al., Ki-67 acts as a biological surfactant to disperse mitotic chromosomes. Nature. (2016)
PubMed: 27362226 DOI: 10.1038/nature18610

Stenström L et al., Mapping the nucleolar proteome reveals a spatiotemporal organization related to intrinsic protein disorder. Mol Syst Biol. (2020)
PubMed: 32744794 DOI: 10.15252/msb.20209469

Derenzini M et al., Nucleolar size indicates the rapidity of cell proliferation in cancer tissues. J Pathol. (2000)
PubMed: 10861579 DOI: 10.1002/(SICI)1096-9896(200006)191:2<181::AID-PATH607>3.0.CO;2-V

Visintin R et al., The nucleolus: the magician's hat for cell cycle tricks. Curr Opin Cell Biol. (2000)
PubMed: 10801456 

Marciniak RA et al., Nucleolar localization of the Werner syndrome protein in human cells. Proc Natl Acad Sci U S A. (1998)
PubMed: 9618508 

Tamanini F et al., The fragile X-related proteins FXR1P and FXR2P contain a functional nucleolar-targeting signal equivalent to the HIV-1 regulatory proteins. Hum Mol Genet. (2000)
PubMed: 10888599 

Willemsen R et al., Association of FMRP with ribosomal precursor particles in the nucleolus. Biochem Biophys Res Commun. (1996)
PubMed: 8769090 DOI: 10.1006/bbrc.1996.1126

Isaac C et al., Characterization of the nucleolar gene product, treacle, in Treacher Collins syndrome. Mol Biol Cell. (2000)
PubMed: 10982400 

Drygin D et al., The RNA polymerase I transcription machinery: an emerging target for the treatment of cancer. Annu Rev Pharmacol Toxicol. (2010)
PubMed: 20055700 DOI: 10.1146/annurev.pharmtox.010909.105844

Spector DL., Macromolecular domains within the cell nucleus. Annu Rev Cell Biol. (1993)
PubMed: 8280462 DOI: 10.1146/annurev.cb.09.110193.001405

Lamond AI et al., Structure and function in the nucleus. Science. (1998)
PubMed: 9554838 

SWIFT H., Studies on nuclear fine structure. Brookhaven Symp Biol. (1959)
PubMed: 13836127 

Lamond AI et al., Nuclear speckles: a model for nuclear organelles. Nat Rev Mol Cell Biol. (2003)
PubMed: 12923522 DOI: 10.1038/nrm1172

Thiry M., The interchromatin granules. Histol Histopathol. (1995)
PubMed: 8573995 

Sleeman JE et al., Newly assembled snRNPs associate with coiled bodies before speckles, suggesting a nuclear snRNP maturation pathway. Curr Biol. (1999)
PubMed: 10531003 

Darzacq X et al., Cajal body-specific small nuclear RNAs: a novel class of 2'-O-methylation and pseudouridylation guide RNAs. EMBO J. (2002)
PubMed: 12032087 DOI: 10.1093/emboj/21.11.2746

Jády BE et al., Modification of Sm small nuclear RNAs occurs in the nucleoplasmic Cajal body following import from the cytoplasm. EMBO J. (2003)
PubMed: 12682020 DOI: 10.1093/emboj/cdg187

Liu Q et al., A novel nuclear structure containing the survival of motor neurons protein. EMBO J. (1996)
PubMed: 8670859 

Lefebvre S et al., Identification and characterization of a spinal muscular atrophy-determining gene. Cell. (1995)
PubMed: 7813012 

Fischer U et al., The SMN-SIP1 complex has an essential role in spliceosomal snRNP biogenesis. Cell. (1997)
PubMed: 9323130 

Lallemand-Breitenbach V et al., PML nuclear bodies. Cold Spring Harb Perspect Biol. (2010)
PubMed: 20452955 DOI: 10.1101/cshperspect.a000661

Booth DG et al., Ki-67 and the Chromosome Periphery Compartment in Mitosis. Trends Cell Biol. (2017)
PubMed: 28838621 DOI: 10.1016/j.tcb.2017.08.001

Ljungberg O et al., A compound follicular-parafollicular cell carcinoma of the thyroid: a new tumor entity? Cancer. (1983)
PubMed: 6136320 DOI: 10.1002/1097-0142(19830915)52:6<1053::aid-cncr2820520621>3.0.co;2-q

Melcák I et al., Nuclear pre-mRNA compartmentalization: trafficking of released transcripts to splicing factor reservoirs. Mol Biol Cell. (2000)
PubMed: 10679009 

Spector DL et al., Associations between distinct pre-mRNA splicing components and the cell nucleus. EMBO J. (1991)
PubMed: 1833187 

Misteli T et al., Protein phosphorylation and the nuclear organization of pre-mRNA splicing. Trends Cell Biol. (1997)
PubMed: 17708924 DOI: 10.1016/S0962-8924(96)20043-1

Cmarko D et al., Ultrastructural analysis of transcription and splicing in the cell nucleus after bromo-UTP microinjection. Mol Biol Cell. (1999)
PubMed: 9880337 

Van Hooser AA et al., The perichromosomal layer. Chromosoma. (2005)
PubMed: 16136320 DOI: 10.1007/s00412-005-0021-9

Booth DG et al., Ki-67 is a PP1-interacting protein that organises the mitotic chromosome periphery. Elife. (2014)
PubMed: 24867636 DOI: 10.7554/eLife.01641

Kau TR et al., Nuclear transport and cancer: from mechanism to intervention. Nat Rev Cancer. (2004)
PubMed: 14732865 DOI: 10.1038/nrc1274

Laurila K et al., Prediction of disease-related mutations affecting protein localization. BMC Genomics. (2009)
PubMed: 19309509 DOI: 10.1186/1471-2164-10-122

Park S et al., Protein localization as a principal feature of the etiology and comorbidity of genetic diseases. Mol Syst Biol. (2011)
PubMed: 21613983 DOI: 10.1038/msb.2011.29

Christoforou A et al., A draft map of the mouse pluripotent stem cell spatial proteome. Nat Commun. (2016)
PubMed: 26754106 DOI: 10.1038/ncomms9992

Itzhak DN et al., Global, quantitative and dynamic mapping of protein subcellular localization. Elife. (2016)
PubMed: 27278775 DOI: 10.7554/eLife.16950

Roux KJ et al., A promiscuous biotin ligase fusion protein identifies proximal and interacting proteins in mammalian cells. J Cell Biol. (2012)
PubMed: 22412018 DOI: 10.1083/jcb.201112098

Lee SY et al., APEX Fingerprinting Reveals the Subcellular Localization of Proteins of Interest. Cell Rep. (2016)
PubMed: 27184847 DOI: 10.1016/j.celrep.2016.04.064

Huh WK et al., Global analysis of protein localization in budding yeast. Nature. (2003)
PubMed: 14562095 DOI: 10.1038/nature02026

Simpson JC et al., Systematic subcellular localization of novel proteins identified by large-scale cDNA sequencing. EMBO Rep. (2000)
PubMed: 11256614 DOI: 10.1093/embo-reports/kvd058

Stadler C et al., Immunofluorescence and fluorescent-protein tagging show high correlation for protein localization in mammalian cells. Nat Methods. 2013 Apr;10(4):315-23 (2013)
PubMed: 23435261 DOI: 10.1038/nmeth.2377

Barbe L et al., Toward a confocal subcellular atlas of the human proteome. Mol Cell Proteomics. (2008)
PubMed: 18029348 DOI: 10.1074/mcp.M700325-MCP200

Stadler C et al., A single fixation protocol for proteome-wide immunofluorescence localization studies. J Proteomics. (2010)
PubMed: 19896565 DOI: 10.1016/j.jprot.2009.10.012

Fagerberg L et al., Mapping the subcellular protein distribution in three human cell lines. J Proteome Res. (2011)
PubMed: 21675716 DOI: 10.1021/pr200379a

Baker M., Reproducibility crisis: Blame it on the antibodies. Nature. (2015)
PubMed: 25993940 DOI: 10.1038/521274a

Jacobson K et al., The Lateral Organization and Mobility of Plasma Membrane Components. Cell. (2019)
PubMed: 31051105 DOI: 10.1016/j.cell.2019.04.018

Kobayashi T et al., Transbilayer lipid asymmetry. Curr Biol. (2018)
PubMed: 29689220 DOI: 10.1016/j.cub.2018.01.007

Krapf D., Compartmentalization of the plasma membrane. Curr Opin Cell Biol. (2018)
PubMed: 29656224 DOI: 10.1016/j.ceb.2018.04.002

Garcia MA et al., Cell-Cell Junctions Organize Structural and Signaling Networks. Cold Spring Harb Perspect Biol. (2018)
PubMed: 28600395 DOI: 10.1101/cshperspect.a029181

Orlando K et al., Membrane organization and dynamics in cell polarity. Cold Spring Harb Perspect Biol. (2009)
PubMed: 20066116 DOI: 10.1101/cshperspect.a001321

Eaton RC et al., D2 receptors in the paraventricular nucleus regulate genital responses and copulation in male rats. Pharmacol Biochem Behav. (1991)
PubMed: 1833780 DOI: 10.1016/0091-3057(91)90418-2

Simons K et al., Cholesterol, lipid rafts, and disease. J Clin Invest. (2002)
PubMed: 12208858 DOI: 10.1172/JCI16390

von Heijne G., Signal sequences. The limits of variation. J Mol Biol. (1985)
PubMed: 4032478 

Johnson AE et al., The translocon: a dynamic gateway at the ER membrane. Annu Rev Cell Dev Biol. (1999)
PubMed: 10611978 DOI: 10.1146/annurev.cellbio.15.1.799

Farhan H et al., Signalling to and from the secretory pathway. J Cell Sci. (2011)
PubMed: 21187344 DOI: 10.1242/jcs.076455

Wishart DS et al., DrugBank: a comprehensive resource for in silico drug discovery and exploration. Nucleic Acids Res. (2006)
PubMed: 16381955 DOI: 10.1093/nar/gkj067

Emanuelsson O et al., Locating proteins in the cell using TargetP, SignalP and related tools. Nat Protoc. (2007)
PubMed: 17446895 DOI: 10.1038/nprot.2007.131

Petersen TN et al., SignalP 4.0: discriminating signal peptides from transmembrane regions. Nat Methods. (2011)
PubMed: 21959131 DOI: 10.1038/nmeth.1701

Käll L et al., A combined transmembrane topology and signal peptide prediction method. J Mol Biol. (2004)
PubMed: 15111065 DOI: 10.1016/j.jmb.2004.03.016

Viklund H et al., SPOCTOPUS: a combined predictor of signal peptides and membrane protein topology. Bioinformatics. (2008)
PubMed: 18945683 DOI: 10.1093/bioinformatics/btn550

Fagerberg L et al., Prediction of the human membrane proteome. Proteomics. (2010)
PubMed: 20175080 DOI: 10.1002/pmic.200900258

Gruenberg J., The endocytic pathway: a mosaic of domains. Nat Rev Mol Cell Biol. (2001)
PubMed: 11584299 DOI: 10.1038/35096054

Griffiths G et al., The mannose 6-phosphate receptor and the biogenesis of lysosomes. Cell. (1988)
PubMed: 2964276 

Schwake M et al., Lysosomal membrane proteins and their central role in physiology. Traffic. (2013)
PubMed: 23387372 DOI: 10.1111/tra.12056

Smith JJ et al., Peroxisomes take shape. Nat Rev Mol Cell Biol. (2013)
PubMed: 24263361 DOI: 10.1038/nrm3700

Greenberg AS et al., Perilipin, a major hormonally regulated adipocyte-specific phosphoprotein associated with the periphery of lipid storage droplets. J Biol Chem. (1991)
PubMed: 2040638 

Walther TC et al., Lipid droplets and cellular lipid metabolism. Annu Rev Biochem. (2012)
PubMed: 22524315 DOI: 10.1146/annurev-biochem-061009-102430

Taguchi T., Emerging roles of recycling endosomes. J Biochem. (2013)
PubMed: 23625997 DOI: 10.1093/jb/mvt034

Bonifacino JS et al., Retrograde transport from endosomes to the trans-Golgi network. Nat Rev Mol Cell Biol. (2006)
PubMed: 16936697 DOI: 10.1038/nrm1985

Antonenkov VD et al., Peroxisomes are oxidative organelles. Antioxid Redox Signal. (2010)
PubMed: 19958170 DOI: 10.1089/ars.2009.2996

Gerst F et al., What role do fat cells play in pancreatic tissue? Mol Metab. (2019)
PubMed: 31113756 DOI: 10.1016/j.molmet.2019.05.001

Applications of Immunocytochemistry - An open-access book about ICC: Ana L. De Paul, J. H. M. J. P. P. S. G. A. A. Q. C. A. M. and A. I. T. Applications of Immunocytochemistry; Dehghani, H., Ed.; InTech, 2012. http://www.intechopen.com/books/applications-of-immunocytochemistry

Immunocytochemistry, a technique for the visualization of proteins and peptides in cells: http://en.wikipedia.org/wiki/Immunocytochemistry

Immunostaining, the use of an antibody-based method to detect a specific target in a sample: http://en.wikipedia.org/wiki/Immunostaining

Immunofluorescence, one type of immunostaining that uses a fluorophore coupled to an antibody for detection: http://en.wikipedia.org/wiki/Immunofluorescence

IHC world – Protocols, Forum, Products, and more: http://www.ihcworld.com/immunocytochemistry.htm

Current Protocols - a continuously updating reference for researchers: http://www.currentprotocols.com/WileyCDA/

The Protocol Exchange - an Open Repository for the deposition and sharing of protocols for scientific research: http://www.nature.com/protocolexchange/protocols

Antibodypedia - An open-access database of publicly available antibodies and their usefulness in various applications: http://www.antibodypedia.com