Nucleoli

Wagner and Valentin were the first to describe the nucleolus in two independent publications in the 1830s. The nucleolus is a nuclear sub-compartment that varies in size and number depending on cell type. The main function of the nucleolus is to synthesize and assemble ribosomes for later transport to the cytoplasm, where translation takes place. The nucleolus is also involved in cell cycle regulation and cellular stress responses. Example images of proteins localized to the nucleoli can be seen in Figure 1.

In the Cell Atlas, 1361 genes (7% of all protein-coding human genes) have been shown to encode proteins that localize to nucleoli (Figure 2). A Gene Ontology (GO)-based functional enrichment analysis of the nucleolar proteins shows enrichment of terms for biological processes related to rRNA processing. Approximately 89% (n=1211) of the nucleolar proteins localize to other cellular compartments in addition to nucleoli, with 34% (n=465) only localizing to other nuclear compartments. The most common additional localization outside the nuclear meta compartment is mitochondria.


UTP6 - A-431

RPF1 - SK-MEL-30

NIFK - A-431

Figure 1. Examples of proteins localized to the nucleoli. UTP6 is suggested to be involved in processing of pre rRNA (detected in A-431 cells). RPF1 is a protein believed to be required for ribosome biogenesis (detected in SK-MEL-30 cells). NIFK is known to localize to the nucleoli, but its function is still unclear (detected in U-2 OS cells).

  • 7% (1361 proteins) of all human proteins have been experimentally detected in the nucleoli by the Human Protein Atlas.
  • 418 proteins in the nucleoli are supported by experimental evidence and out of these 122 proteins are enhanced by the Human Protein Atlas.
  • 1211 proteins in the nucleoli have multiple locations.
  • 339 proteins in the nucleoli show a cell to cell variation. Of these 295 show a variation in intensity and 55 a spatial variation.

  • Nucleolar proteins are mainly involved in rRNA processing.

Figure 2. 7% of all human protein-coding genes encode proteins localized to the nucleoli. Each bar is clickable and gives a search result of proteins that belong to the selected category.

The structure of nucleoli

Substructures

  • Nucleoli: 1008
  • Nucleoli fibrillar center: 300
  • Nucleoli rim: 159

The nucleoli are non-membrane enclosed, highly conserved, sub-organelles within the nucleus. They are formed around nucleolus organizer regions (NORs) consisting of ribosomal DNA (rDNA) and are structurally organized into three different sub regions; the fibrillar center (FC), the dense fibrillar component (DFC) and the granular component (GC) (Boisvert FM et al. (2007); Scheer U et al. (1999)). A selection of proteins localized to the nucleoli that are suitable as nucleoli markers can be found in Table 1.

Table 1. Selection of proteins suitable as markers for the nucleoli or its substructures.

Gene Description Substructure
DDX47 DEAD-box helicase 47 Nucleoli
RPF1 Ribosome production factor 1 homolog Nucleoli
UTP6 UTP6, small subunit processome component Mitotic chromosome
Nucleoli
NOL10 Nucleolar protein 10 Nucleoli
FTSJ3 FtsJ RNA methyltransferase homolog 3 Mitotic chromosome
Nucleoli
Nucleoli rim
UBTF Upstream binding transcription factor, RNA polymerase I Nucleoli fibrillar center

A majority of the nucleolar proteins show staining throughout the whole nucleolar area, while roughly 20% display a more refined staining pattern. The staining of fibrillar centers and/or dense fibrillar component appears as clusters of spots for most cell lines while for others, for example MCF-7 and U-251, only one larger spot is seen. Some proteins localize to the rim of the nucleolus, which is visible as a thin circle around the nucleolus and could be associated to either the GC or the perinucleolar heterochromatin surrounding the nucleolus (Németh A et al. (2011)). A recent study suggests that the protein MKI67, which is localized to the nucleoli rim, functions like a surfactant to create non-membranous barriers in the cell. Therefore, proteins with similar staining patterns could have a similar function (Cuylen S et al. (2016); Stenström L et al. (2020)). MKI67 and other immunofluorescent images of different nucleolar substructures can be seen in Figure 3. The size of the nucleolus has also been suggested to correlate with the proliferative ability of cells (Derenzini M et al. (2000)). Upon entry into mitosis, rRNA transcription and RNP processing shuts down and the nucleoli are disassembled. In telophase and early G1, the nucleolar organization is re-established.


KRI1 - HEK 293

KRI1 - MCF7

KRI1 - U-2 OS


NOLC1 - HEK 293

UBTF - U-2 OS

MKI67 - U-251 MG

Figure 3. Examples of the morphology of the nucleoli in different cell lines as well as the nucleolar substructures and staining patterns. Immunofluorescent staining of KRI1 in HEK 239, MCF-7 and U-2 OS cells. NOLC1 might play a role in maintaining the structure of the fibrillar center and the dense fibrillar component in the nucleoli. NOLC1 is localized to the fibrillar center (detected in HEK293 cells). UBTF is involved in the activation of RNA polymerase I and is localized to the fibrillar center (detected in U-2 OS cells). MKI67 has been found to maintain mitotic chromosome integrity and is a well-known cellular proliferation marker.


Figure 4. 3D-view of nucleoli in U-2 OS, visualized by immunofluorescent staining of NOP56. The morphology of nucleoli in human induced stem cells can be seen in the Allen Cell Explorer.

The function of nucleoli

The nucleolus is responsible for the synthesis, processing and assembly of ribosomes, a complex process controlled in the nucleolar sub regions (Boisvert FM et al. (2007); Scheer U et al. (1999); Németh A et al. (2011)). The border between the FC and the DFC contains proteins from the RNA polymerase I complex and is the region where pre-ribosomal RNA (pre-rRNA) is transcribed from rDNA. The pre-rRNA is later modified by proteins in the DFC followed by assembly of the ribosome subunits in the GC (Scheer U et al. (1999)). As is the case for the majority of organelles, the proteome of the nucleolus is dynamic and has been shown to consist of multiple overlapping sets of proteins that are interchanging dependent on the cell state. The need for translational capacity varies with different cell cycle phases, and transcriptional capacity is heavily dependent on the amount of ribosomes available. In addition to being responsible for ribosome assembly, the nucleolus has also been found to comprise proteins involved in cell cycle regulation and cellular stress responses (Boisvert FM et al. (2007); Visintin R et al. (2000)).

Several genetic disorders such as Werner syndrome, fragile X syndrome and Treacher Collins syndrome have been linked to nucleolar proteins (Marciniak RA et al. (1998); Tamanini F et al. (2000); Willemsen R et al. (1996); Isaac C et al. (2000)). Moreover, the nucleolar size increases with the proliferative ability of cells, suggesting that the nucleoli play an important role in development of cancer and could therefore be a potential target for cancer therapy (Drygin D et al. (2010)).

Gene Ontology (GO) analysis of the proteins mainly localized to the nucleoli shows functions that are well in-line with already known functions for the structure. The enriched terms for the GO domain Biological Process are related to rRNA processing and ribosome assembly (Figure 5a), while enrichment analysis of the GO domain Molecular Function gives enrichment for RNA binding activities (Figure 5b). A list of highly expressed nucleolar proteins are summarized in Table 2.

Figure 5a. Gene Ontology-based enrichment analysis for the nucleolar proteome showing the significantly enriched terms for the GO domain Biological Process. Each bar is clickable and gives a search result of proteins that belong to the selected category.

Figure 5b. Gene Ontology-based enrichment analysis for the nucleolar proteome showing the significantly enriched terms for the GO domain Molecular Function. Each bar is clickable and gives a search result of proteins that belong to the selected category.

Table 2. Highly expressed single localized nucleolar proteins across different cell lines.

Gene Description Average NX
FBL Fibrillarin 34
NOLC1 Nucleolar and coiled-body phosphoprotein 1 29
POLR2K RNA polymerase II subunit K 29
NOP56 NOP56 ribonucleoprotein 28
WDR82 WD repeat domain 82 25
ZNF146 Zinc finger protein 146 24
UBTF Upstream binding transcription factor, RNA polymerase I 23
NOP53 NOP53 ribosome biogenesis factor 22
NOP16 NOP16 nucleolar protein 22
RPF1 Ribosome production factor 1 homolog 22

Nucleolar proteins with multiple locations

Of the nucleolar proteins identified in the Cell Atlas, approximately 89% (n=1211) also localize to other cell compartments (Figure 6). 34% (n=465) of the nucleolar proteins only localize to other nuclear structures. The network plot shows that the most common locations shared with nucleoli are nucleoplasm, cytosol and mitochondria. Given that the nucleoli are responsible for synthesis and assembly of ribosomes that later are exported to the cytoplasm, many of the proteins localized to both the nucleoli and the cytoplasmic structures are most likely involved in translation. The number of proteins localized to the nucleoli and the nucleoplasm as well as the nucleoli and mitochondria are seen more often than expected with the current distribution of multilocalizing proteins, while nucleolar proteins that additionally localize to vesicles, the Golgi apparatus, the cytosol or the centrosomes are significantly underrepresented. Examples of multilocalizing proteins within the nucleolar proteome can be seen in Figure 7.

Figure 6. Interactive network plot of nucleolar proteins with multiple localizations. The numbers in the connecting nodes show the proteins that are localized to the nucleoli and to one or more additional locations. Only connecting nodes containing more than one protein and at least 0.5% of proteins in the nucleolar proteome are shown. The circle sizes are related to the number of proteins. The cyan colored nodes show combinations that are significantly overrepresented, while magenta colored nodes show combinations that are significantly underrepresented as compared to the probability of observing that combination based on the frequency of each annotation and a hypergeometric test (p≤0.05). Note that this calculation is only done for proteins with dual localizations. Each node is clickable and results in a list of all proteins that are found in the connected organelles.


EXOSC10 - U-2 OS

APTX - PC-3

SPATS2L - U-2 OS

Figure 7. Examples of multilocalizing proteins in the nucleolar proteome. The examples show common or overrepresented combinations for multilocalizing proteins in the nucleolar proteome. EXOSC10 is known to be involved in multiple RNA processing pathways in the nucleolus, nucleus and the cytoplasm (detected in U-2 OS cells). APTX is known to be involved in DNA repair and is localized to the nucleoplasm and the nucleoli (detected in PC-3 cells). SPATS2L is known to localize to the nucleoli and into cytoplasmic stress granules during oxidative stress but the function is unknown (detected in U-2 OS cells).

Expression levels of nucleoli proteins in tissue

Transcriptome analysis and classification of genes into tissue distribution categories (Figure 8) shows that genes encoding proteins that localize to nucleoli are more likely to either be detected in a single tissue or detected in all tissues, compared to all genes presented in the Cell Atlas. Significantly lower portions of nucleoli-associated genes are detected in some or many of the tissues. Thus, these genes tend to either be ubiquitously expressed, or show a strict tissue-specific expression.

Figure 8. Bar plot showing the percentage of genes in different tissue distribution categories for nucleoli-associated protein-coding genes compared to all genes in the Cell Atlas. Asterisk marks a statistically significant deviation (p≤0.05) in the number of genes in a category based on a binomial statistical test. Each bar is clickable and gives a search result of proteins that belong to the selected category.

Relevant links and publications

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