Mitochondria

Mitochondria generate the energy that is needed to power the functions of the cell, but they also participate directly in several other cellular processes, including apoptosis, cell cycle control and calcium homeostasis. Mitochondria are distributed throughout the cytoplasm and vary in number between different cell types. Each organelle is enclosed by a double membrane, with the inner one forming the characteristic folds known as cristae. Mutations causing mitochondrial dysfunction are often related to severe diseases. Examples of proteins localized to mitochondria can be seen in Figure 1.

In the subcellular section, 1121 genes (6% of all protein-coding human genes) have been shown to encode proteins that localize to mitochondria (Figure 2). A Gene Ontology (GO)-based enrichment analysis of genes encoding mitochondrial proteins shows en enrichment of genes associated with biological processes related to cellular respiration as well as to mitochondrial organization, gene expression and metabolic processes. Approximately 48% (n=542) of the mitochondrial proteome localizes to additional cellular compartments, most commonly to the nucleoplasm, nucleoli and/or the cytosol.


LRPPRC - U2OS

CHCHD3 - U2OS

CS - U2OS


PHB2 - U2OS

TRAP1 - U2OS

IMMT - U2OS


PCK2 - A-431

PYCR2 - U-251MG

PGAM5 - HEK293

Figure 1. Examples of proteins localized to the mitochondria. LRPPRC might play a role in transcription of mitochondrial genes (detected in U2OS cells). CHCHD3 is a protein in the MICOS complex, localized to the mitochondrial inner membrane (detected in U2OS cells). CS is active in the citric acid cycle (detected in U2OS cells). PHB2 is probably involved in the regulation of mitochondrial respiration activity (detected in U2OS cells). TRAP1 is important for maintaining mitochondrial function and polarization (detected in U2OS cells). IMMT is, just like CHCHD3, part of the MICOS complex located in the inner membrane (detected in U2OS cells). PCK2 catalyzes the conversion of oxaloacetate to phosphoenolpyruvate (detected in A-431 cells). PYCR2 catalyzes the last step in proline biosynthesis (detected in U-251 MG cells). PGAM5 may be a regulator of mitochondrial dynamics (detected in HEK 293 cells).

  • 6% (1121 proteins) of all human proteins have been experimentally detected in the mitochondria by the Human Protein Atlas.
  • 515 proteins in the mitochondria are supported by experimental evidence and out of these 119 proteins are enhanced by the Human Protein Atlas.
  • 542 proteins in the mitochondria have multiple locations.
  • 343 proteins in the mitochondria show single cell variation.

  • Mitochondrial proteins are mainly involved in cellular respiration and in mitochondrial organization, gene expression and metabolic processes.

Figure 2. 6% of all human protein-coding genes encode proteins localized to the mitochondria. Each bar is clickable and gives a search result of proteins that belong to the selected category.

The structure of mitochondria

The mitochondrion was first described in 1890 by Richard Altmann. It is approximately 0.5-1 μm long and enclosed by an outer and inner membrane seprated by an intermembrane space. The folds of the inner membrane, denoted cristae, enclose the aqueous matrix, which contains the mitochondrial DNA (mtDNA) and the majority of the mitochondrial proteins (Nunnari J et al. (2012)). The mitochondrion is the only organelle in animals to possess a small genome of its own, consisting of 37 genes in a circular genome of exclusively maternal inheritance. Of these genes, 13 encode proteins in the respiratory chain, 22 encode transfer RNAs and 2 encode mitochondrial ribosomal RNAs (Friedman JR et al. (2014). However, the mitochondrial proteome has been estimated to contain around 1000-1500 proteins, and thus the great majority are encoded by nuclear genes and imported into mitochondria (Nunnari J et al. (2012); Nunnari J et al. (2012); Friedman JR et al. (2014); Calvo SE et al. (2010). Table 1 contains a list of proteins suitable as markers for mitochondria, while Table 2 contains highly expressed genes that encode mitochondrial proteins.

Table 1. Selection of proteins suitable as markers for mitochondria.

Gene Description Substructure
CS Citrate synthase Mitochondria
LRPPRC Leucine rich pentatricopeptide repeat containing Mitochondria
SLC25A24 Solute carrier family 25 member 24 Mitochondria
TIMM44 Translocase of inner mitochondrial membrane 44 Mitochondria
GCDH Glutaryl-CoA dehydrogenase Mitochondria
TRAP1 TNF receptor associated protein 1 Mitochondria

Table 2. Highly expressed single localizing mitochondrial proteins across different cell lines.

Gene Description Average nTPM
MT-CO1 Mitochondrially encoded cytochrome c oxidase I 11707
MT-ND4 Mitochondrially encoded NADH:ubiquinone oxidoreductase core subunit 4 6512
MT-CYB Mitochondrially encoded cytochrome b 3734
ATP5F1B ATP synthase F1 subunit beta 892
HSPD1 Heat shock protein family D (Hsp60) member 1 742
CHCHD2 Coiled-coil-helix-coiled-coil-helix domain containing 2 695
COX6B1 Cytochrome c oxidase subunit 6B1 678
SLC25A3 Solute carrier family 25 member 3 673
COX4I1 Cytochrome c oxidase subunit 4I1 654
ATP5ME ATP synthase membrane subunit e 628

The number of mitochondria varies with cell type and according to the energy needs of individual cells. Mitochondria are continuously undergoing fission and fusion, which allows for regulation of the number of mitochondria as well as communication and exchange of mitochondrial components. Loss of mitochondrial fission/fusion function is associated with defects in oxidative phosphorylation and loss of mtDNA (Friedman JR et al. (2014)). The morphology and distribution of mitochondria varies between different cell types, as shown in the examples of Figure 3.


ALDH5A1 - CACO-2

NIPSNAP2 - SH-SY5Y

NDUFAF2 - MCF-7


PCK2 - Hep-G2

MAOA - RT-4

SDHA - HeLa

Figure 3. Examples of the morphology of mitochondria in different cell lines, represented by immunofluorescent staining of different mitochondrial proteins. ALDH5A1 in CACO-2 cells, NIPSNAP2 in SH-SY5Y cells and NDUFAF2 in MCF-7 cells. PCK2 in Hep-G2 cells, MAOA in RT-4 cells and SDHA in HeLa cells.


Figure 4. 3D-view of mitochondria in U2OS, visualized by immunofluorescent staining of CS. The morphology of mitochondria in human induced stem cells can be seen in the Allen Cell Explorer.

The function of mitochondria

Mitochondria are well-known for their function in generating ATP through the electron transport chain and ATP synthase, which is located in the inner membrane, in a process known as oxidative phosphorylation. However, mitochondria are also involved in several other cellular processes, including regulation of metabolism, calcium homeostasis and cell signaling (McBride HM et al. (2006)). They also have an important role in cell cycle control, cell growth, differentiation, and apoptosis.

The incidence of mitochondrial disorders has been estimated to 1 in 5000 individuals or higher, making it one of the most common types of heritable human diseases (Schaefer AM et al. (2004)). These disorders can be caused by mutations in mitochondrial and/or nuclear DNA, and phenotypically different diseases may stem from mutations in the same protein complexes (Nunnari J et al. (2012)).

Gene Ontology (GO)-based analysis of genes encoding proteins that localize to mitochondria shows enrichment of terms that are well in-line with the known functions of mitochondria. The most highly enriched terms for the GO domain Biological Process are related to transcription, translation and processing of proteins in mitochondria, structural organization of mitochondria, cellular respiration and metabolic processes (Figure 5a). Enrichment analysis of the GO domain Molecular Function also shows significant enrichment for terms related to energy production, such as NADH dehydrogenase and oxidoreductase activity, as well as transmembrane transporter activity (Figure 5b).

Figure 5a. Gene Ontology-based enrichment analysis for the mitochondria 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 mitochondria 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.

Mitochondria proteins with multiple locations

Among the mitochondrial proteins detected in the subcellular section, 48% (n=542) also localize to other cellular compartments (Figure 6). The network plot shows that the most common locations shared with mitochondria are nucleoplasm, cytosol and nucleoli, with proteins localizing to mitochondria and nucleoplasm or nucleoli being overrepresented. Localization to both mitochondria and nucleus could highlight proteins functioning in gene expression, which occurs in both of these compartments. Examples of mitochondrial proteins also localizing to other cellular compartments are shown in Figure 7.

Figure 6. Interactive network plot of mitochondrial proteins with multiple localizations. The numbers in the connecting nodes show the proteins that are localized to the mitochondria and to one or more additional locations. Only connecting nodes containing more than one protein and at least 0.5% of proteins in the mitochondrial 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.


CCDC51 - U2OS

FAM162A - U-251MG

COX7A2L - PC-3

Figure 7. Examples of multilocalizing proteins in the mitochondrial proteome. The examples show common or overrepresented combinations for multilocalizing proteins in the mitochondrial proteome. CCDC51 is an uncharacterized protein localized to both the nucleoplasm and mitochondria (detected in U2OS cells). FAM162A has been proposed to be involved in regulation of apoptosis (detected in U-251 MG cells). COX7A2L is an uncharacterized protein (detected in PC-3 cells).

Expression levels of mitochondria proteins in tissue

Transcriptome analysis and classification of genes into tissue distribution categories (Figure 8) shows that a larger portion of genes encoding mitochondrial proteins are detected in all tissues, while smaller portions are detected in some or many tissues, compared to all genes presented in the subcellular section. There is also a significantly smaller portion of the genes encoding proteins that localize to mitochondria that are not detected in any of the tissues that have been analyzed. This is in agreement with the roles of mitochondria in basic and essential functions in all cells of the human body.

Figure 8. Bar plot showing the percentage of genes in different tissue distribution categories for mitochondria-associated protein-coding genes, compared to all genes in the subcellular section. 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

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

Hildreth AD et al., Single-cell sequencing of human white adipose tissue identifies new cell states in health and obesity. Nat Immunol. (2021)
PubMed: 33907320 DOI: 10.1038/s41590-021-00922-4

He S et al., Single-cell transcriptome profiling of an adult human cell atlas of 15 major organs. Genome Biol. (2020)
PubMed: 33287869 DOI: 10.1186/s13059-020-02210-0

Bhat-Nakshatri P et al., A single-cell atlas of the healthy breast tissues reveals clinically relevant clusters of breast epithelial cells. Cell Rep Med. (2021)
PubMed: 33763657 DOI: 10.1016/j.xcrm.2021.100219

Lukassen S et al., SARS-CoV-2 receptor ACE2 and TMPRSS2 are primarily expressed in bronchial transient secretory cells. EMBO J. (2020)
PubMed: 32246845 DOI: 10.15252/embj.20105114

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

Wang W et al., Single-cell transcriptomic atlas of the human endometrium during the menstrual cycle. Nat Med. (2020)
PubMed: 32929266 DOI: 10.1038/s41591-020-1040-z

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

Ulrich ND et al., Cellular heterogeneity of human fallopian tubes in normal and hydrosalpinx disease states identified using scRNA-seq. Dev Cell. (2022)
PubMed: 35320732 DOI: 10.1016/j.devcel.2022.02.017

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

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

Tabula Sapiens Consortium* et al., The Tabula Sapiens: A multiple-organ, single-cell transcriptomic atlas of humans. Science. (2022)
PubMed: 35549404 DOI: 10.1126/science.abl4896

Wagner M et al., Single-cell analysis of human ovarian cortex identifies distinct cell populations but no oogonial stem cells. Nat Commun. (2020)
PubMed: 32123174 DOI: 10.1038/s41467-020-14936-3

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

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

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

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

De Micheli AJ et al., A reference single-cell transcriptomic atlas of human skeletal muscle tissue reveals bifurcated muscle stem cell populations. Skelet Muscle. (2020)
PubMed: 32624006 DOI: 10.1186/s13395-020-00236-3

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

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

Agaton C et al., Affinity proteomics for systematic protein profiling of chromosome 21 gene products in human tissues. Mol Cell Proteomics. (2003)
PubMed: 12796447 DOI: 10.1074/mcp.M300022-MCP200

Lindskog M et al., Selection of protein epitopes for antibody production Biotechniques (2005)
PubMed: 15945371 

Larsson M et al., High-throughput protein expression of cDNA products as a tool in functional genomics. J Biotechnol. (2000)
PubMed: 10908795 DOI: 10.1016/s0168-1656(00)00258-3

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

Uhlén M et al., The human secretome. Sci Signal. (2019)
PubMed: 31772123 DOI: 10.1126/scisignal.aaz0274

Uhlen M et al., A genome-wide transcriptomic analysis of protein-coding genes in human blood cells. Science. (2019)
PubMed: 31857451 DOI: 10.1126/science.aax9198

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

Zhong W et al., The neuropeptide landscape of human prefrontal cortex. Proc Natl Acad Sci U S A. (2022)
PubMed: 35947618 DOI: 10.1073/pnas.2123146119

Sjöstedt E et al., An atlas of the protein-coding genes in the human, pig, and mouse brain. Science. (2020)
PubMed: 32139519 DOI: 10.1126/science.aay5947

Gilvesy A et al., Spatiotemporal characterization of cellular tau pathology in the human locus coeruleus-pericoerulear complex by three-dimensional imaging. Acta Neuropathol. (2022)
PubMed: 36040521 DOI: 10.1007/s00401-022-02477-6

Jin H et al., Systematic transcriptional analysis of human cell lines for gene expression landscape and tumor representation. Nat Commun. (2023)
PubMed: 37669926 DOI: 10.1038/s41467-023-41132-w

Schubert M et al., Perturbation-response genes reveal signaling footprints in cancer gene expression. Nat Commun. (2018)
PubMed: 29295995 DOI: 10.1038/s41467-017-02391-6

Jiang P et al., Systematic investigation of cytokine signaling activity at the tissue and single-cell levels. Nat Methods. (2021)
PubMed: 34594031 DOI: 10.1038/s41592-021-01274-5

Jin L et al., Targeting of CD44 eradicates human acute myeloid leukemic stem cells. Nat Med. (2006)
PubMed: 16998484 DOI: 10.1038/nm1483

Magis AT et al., Untargeted longitudinal analysis of a wellness cohort identifies markers of metastatic cancer years prior to diagnosis. Sci Rep. (2020)
PubMed: 33004987 DOI: 10.1038/s41598-020-73451-z

Santarius T et al., GLO1-A novel amplified gene in human cancer. Genes Chromosomes Cancer. (2010)
PubMed: 20544845 DOI: 10.1002/gcc.20784

Berggrund M et al., Identification of Candidate Plasma Protein Biomarkers for Cervical Cancer Using the Multiplex Proximity Extension Assay. Mol Cell Proteomics. (2019)
PubMed: 30692274 DOI: 10.1074/mcp.RA118.001208

Virgilio L et al., Deregulated expression of TCL1 causes T cell leukemia in mice. Proc Natl Acad Sci U S A. (1998)
PubMed: 9520462 DOI: 10.1073/pnas.95.7.3885

Saberi Hosnijeh F et al., Proteomic markers with prognostic impact on outcome of chronic lymphocytic leukemia patients under chemo-immunotherapy: results from the HOVON 109 study. Exp Hematol. (2020)
PubMed: 32781097 DOI: 10.1016/j.exphem.2020.08.002

Gao L et al., Integrative analysis the characterization of peroxiredoxins in pan-cancer. Cancer Cell Int. (2021)
PubMed: 34246267 DOI: 10.1186/s12935-021-02064-x

Satelli A et al., Galectin-4 functions as a tumor suppressor of human colorectal cancer. Int J Cancer. (2011)
PubMed: 21064109 DOI: 10.1002/ijc.25750

Harlid S et al., A two-tiered targeted proteomics approach to identify pre-diagnostic biomarkers of colorectal cancer risk. Sci Rep. (2021)
PubMed: 33664295 DOI: 10.1038/s41598-021-83968-6

Sun X et al., Prospective Proteomic Study Identifies Potential Circulating Protein Biomarkers for Colorectal Cancer Risk. Cancers (Basel). (2022)
PubMed: 35805033 DOI: 10.3390/cancers14133261

Bhardwaj M et al., Comparison of Proteomic Technologies for Blood-Based Detection of Colorectal Cancer. Int J Mol Sci. (2021)
PubMed: 33530402 DOI: 10.3390/ijms22031189

Chen H et al., Head-to-Head Comparison and Evaluation of 92 Plasma Protein Biomarkers for Early Detection of Colorectal Cancer in a True Screening Setting. Clin Cancer Res. (2015)
PubMed: 26015516 DOI: 10.1158/1078-0432.CCR-14-3051

Thorsen SB et al., Detection of serological biomarkers by proximity extension assay for detection of colorectal neoplasias in symptomatic individuals. J Transl Med. (2013)
PubMed: 24107468 DOI: 10.1186/1479-5876-11-253

Mahboob S et al., A novel multiplexed immunoassay identifies CEA, IL-8 and prolactin as prospective markers for Dukes' stages A-D colorectal cancers. Clin Proteomics. (2015)
PubMed: 25987887 DOI: 10.1186/s12014-015-9081-x

He W et al., Attenuation of TNFSF10/TRAIL-induced apoptosis by an autophagic survival pathway involving TRAF2- and RIPK1/RIP1-mediated MAPK8/JNK activation. Autophagy. (2012)
PubMed: 23051914 DOI: 10.4161/auto.22145

Enroth S et al., A two-step strategy for identification of plasma protein biomarkers for endometrial and ovarian cancer. Clin Proteomics. (2018)
PubMed: 30519148 DOI: 10.1186/s12014-018-9216-y

Jung CS et al., Serum GFAP is a diagnostic marker for glioblastoma multiforme. Brain. (2007)
PubMed: 17998256 DOI: 10.1093/brain/awm263

Jaworski DM et al., BEHAB (brain enriched hyaluronan binding) is expressed in surgical samples of glioma and in intracranial grafts of invasive glioma cell lines. Cancer Res. (1996)
PubMed: 8625302 

Zhang X et al., CEACAM5 stimulates the progression of non-small-cell lung cancer by promoting cell proliferation and migration. J Int Med Res. (2020)
PubMed: 32993395 DOI: 10.1177/0300060520959478

Xu F et al., A Linear Discriminant Analysis Model Based on the Changes of 7 Proteins in Plasma Predicts Response to Anlotinib Therapy in Advanced Non-Small Cell Lung Cancer Patients. Front Oncol. (2021)
PubMed: 35070967 DOI: 10.3389/fonc.2021.756902

Dagnino S et al., Prospective Identification of Elevated Circulating CDCP1 in Patients Years before Onset of Lung Cancer. Cancer Res. (2021)
PubMed: 33574093 DOI: 10.1158/0008-5472.CAN-20-3454

Álvez MB et al., Next generation pan-cancer blood proteome profiling using proximity extension assay. Nat Commun. (2023)
PubMed: 37463882 DOI: 10.1038/s41467-023-39765-y

Wik L et al., Proximity Extension Assay in Combination with Next-Generation Sequencing for High-throughput Proteome-wide Analysis. Mol Cell Proteomics. (2021)
PubMed: 34715355 DOI: 10.1016/j.mcpro.2021.100168

Zeiler M et al., A Protein Epitope Signature Tag (PrEST) library allows SILAC-based absolute quantification and multiplexed determination of protein copy numbers in cell lines. Mol Cell Proteomics. (2012)
PubMed: 21964433 DOI: 10.1074/mcp.O111.009613

Peng Y et al., Identification of key biomarkers associated with cell adhesion in multiple myeloma by integrated bioinformatics analysis. Cancer Cell Int. (2020)
PubMed: 32581652 DOI: 10.1186/s12935-020-01355-z

Gyllensten U et al., Next Generation Plasma Proteomics Identifies High-Precision Biomarker Candidates for Ovarian Cancer. Cancers (Basel). (2022)
PubMed: 35406529 DOI: 10.3390/cancers14071757

Enroth S et al., High throughput proteomics identifies a high-accuracy 11 plasma protein biomarker signature for ovarian cancer. Commun Biol. (2019)
PubMed: 31240259 DOI: 10.1038/s42003-019-0464-9

Wang Z et al., DNER promotes epithelial-mesenchymal transition and prevents chemosensitivity through the Wnt/β-catenin pathway in breast cancer. Cell Death Dis. (2020)
PubMed: 32811806 DOI: 10.1038/s41419-020-02903-1

Liu S et al., Discovery of CASP8 as a potential biomarker for high-risk prostate cancer through a high-multiplex immunoassay. Sci Rep. (2021)
PubMed: 33828176 DOI: 10.1038/s41598-021-87155-5

Orchard S et al., The MIntAct project--IntAct as a common curation platform for 11 molecular interaction databases. Nucleic Acids Res. (2014)
PubMed: 24234451 DOI: 10.1093/nar/gkt1115

Robinson JL et al., An atlas of human metabolism. Sci Signal. (2020)
PubMed: 32209698 DOI: 10.1126/scisignal.aaz1482

Uhlen M et al., A pathology atlas of the human cancer transcriptome. Science. (2017)
PubMed: 28818916 DOI: 10.1126/science.aan2507

Hikmet F et al., The protein expression profile of ACE2 in human tissues. Mol Syst Biol. (2020)
PubMed: 32715618 DOI: 10.15252/msb.20209610

Gordon DE et al., A SARS-CoV-2 protein interaction map reveals targets for drug repurposing. Nature. (2020)
PubMed: 32353859 DOI: 10.1038/s41586-020-2286-9

Karlsson M et al., A single-cell type transcriptomics map of human tissues. Sci Adv. (2021)
PubMed: 34321199 DOI: 10.1126/sciadv.abh2169

Jumper J et al., Highly accurate protein structure prediction with AlphaFold. Nature. (2021)
PubMed: 34265844 DOI: 10.1038/s41586-021-03819-2

Varadi M et al., AlphaFold Protein Structure Database: massively expanding the structural coverage of protein-sequence space with high-accuracy models. Nucleic Acids Res. (2022)
PubMed: 34791371 DOI: 10.1093/nar/gkab1061

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

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

Mahdessian D et al., Spatiotemporal dissection of the cell cycle with single-cell proteogenomics. Nature. (2021)
PubMed: 33627808 DOI: 10.1038/s41586-021-03232-9

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

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 

Becht E et al., Dimensionality reduction for visualizing single-cell data using UMAP. Nat Biotechnol. (2018)
PubMed: 30531897 DOI: 10.1038/nbt.4314

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

Yoon TY et al., SNARE complex assembly and disassembly. Curr Biol. (2018)
PubMed: 29689222 DOI: 10.1016/j.cub.2018.01.005

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

Ouyang W et al., Analysis of the Human Protein Atlas Image Classification competition. Nat Methods. (2019)
PubMed: 31780840 DOI: 10.1038/s41592-019-0658-6

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