Sperm
The sperm cell, or spermatozoon, is the male motile gamete of humans and animals. It was first described in 1677 by van Leeuwenhoek. In contrast to other cell types, spermatozoa lacks most of the standard organelles, such as the endoplasmic reticulum and the Golgi apparatus. During spermatogenesis, stem cells in the testis develop into a highly specialized haploid cell type that is optimized for swimming to, penetrating, and ultimately fusing with the female gamete, known as the oocyte, to form a diploid and totipotent zygote. To achieve this, sperm cells contain several unique subcellular structures, including a long motile cilium called the flagellum, allowing high motility and aiding egg penetration, and a sac containing enzymes used for penetrating the egg, called the acrosome. Examples of proteins localizing to different regions of spermatozoa can be seen in Figure 1.
In the subcellular resource, 645 genes (3% of all human protein-coding genes) have been shown to encode proteins that localize to human spermatozoa and their unique sub-compartments (Figure 2). There are also 65 genes encoding proteins that have been found to loalize to mitochondria in sperm. A Gene Ontology (GO)-based functional enrichment analysis of genes encoding proteins in the sperm-specific substructures shows an enrichment of genes associated with biological processes related to sperm motility, organization and assembly of the cilium, spermatogenesis, and microtubule-based transport. Approximately 76% (n=493) of the proteins that localize to the sperm cell can also be detected in additional cellular compartments in other cell types, with the most common additional localization being mitochondria.
SPACA1 - Sperm
DYDC1 - Sperm
ARMC12 - Sperm
Figure 1. Examples of proteins localized to the sperm cell and some of its substructures. SPACA1 is a key protein for establishing a normal morphology during spermatogenesis and plays a role in the formation of the acrosome vesicle. DYDC1 localizes along the length of the entire flagellum and is important for sperm motility. ARMC12 is a sperm mitochondrial protein that is essential for male fertility and for the formation of the mitochondrial sheath in the mid piece of sperm cells.
- 3% (645 proteins) of all human proteins have been experimentally detected in the sperm by the Human Protein Atlas.
- 73 proteins in the sperm are supported by experimental evidence and out of these 3 proteins are enhanced by the Human Protein Atlas.
- 493 proteins in the sperm have multiple locations.
- 236 proteins in the sperm show single cell variation.
- Proteins localizing to the sperm cell are mainly involved in spermatogenesis, sperm motility, cilium assembly and organization, and microtubule-based transport.
Figure 2. 3% of all human protein-coding genes encode proteins that have been shown to localize to the sperm cell and its substructures. Each bar is clickable and gives a search result of proteins that belong to the selected category.
The structure of the sperm cell
Substructures
- Acrosome: 133
- Equatorial segment: 81
- Perinuclear theca: 68
- Calyx: 70
- Connecting piece: 93
- Flagellar centriole: 115
- Mid piece: 350
- Principal piece: 387
- End piece: 194
- Annulus: 50
The sperm cell is a highly specialized cell type with a distinct morphology compared to other cell types. During spermatogenesis, stem cells in testis undergo several steps of morphological and functional remodeling, leading to the formation of a haploid cell, consisting of a head region with species-specific morphology connected to a long tail (Schneider S et al. (2023)).
The head region of sperm contains the nucleus, the acrosome, the equatorial segment, and the perinuclear theca, including the calyx. The nucleus contains the haploid paternal genome, with one copy of each chromosome, as well as a multitude of different RNA molecules. The DNA in the nucleus of the sperm head is hypercondensed into a genetically inactive state, with protamines substituting the core histones. The acrosome covers the anterior portion of the sperm head and is derived from the Golgi apparatus as well as the plasma membrane of sperm progenitor cells. It contains different digestive enzymes and other molecules that are exocytosed during the acrosome reaction, which occurs when the sperm cell touches the egg coat and which is essential to enable penetration and fertilization. The equatorial segment is a region in the middle portion of the sperm head that contains a unique set of proteins, which seem to play a role in plasmamembrane fusion between the spermatozoon and the oocyte. The condensed cytosolic regions, or perinuclear theca, of the sperm head can been divided into a subacrosomal and postacrosomal part. The subacrosomal part, referred to as the perinuclear theca in the sucelluar resource, develops early during spermatogenesis and is a thin cytosolic layer between the acrosome vesicle and nuclear envelope, which is continuous with the outer periacrosomal layer. This region is important for acrosome assebly. The postacrosomal part, known as the calyx, is localized between the nuclear envelope and the sperm plasma membrane. It consists of cytosolic proteins that are transported via the sperm manchette and is formed during the maturation phase when the sperm head is elongated, and is important for sperm–egg interactions and egg activation during fertilization (Balhorn R. (2007); Khawar MB et al. (2019); Schneider S et al. (2023)). Examples of proteins localizing to the sperm head regions can be seen in Figure 3.
SPACA3 - Acrosome
SPESP1 - Equatorial segment
RAP1GAP2 - Perinuclear theca
CYLC2 - Calyx
Figure 3. Examples showing the different sperm head substructures and staining patterns. SPACA3 is localized to the acrosome and is thought to be involved in the sperm-egg fusion event during fertilization. SPESP1 is localized to the equatorial segment and is important for the fertilization ability of the sperm cell. RAP1GAP2 is localized to the perinuclear theca and is involed in the acrosome reaction. CYLC2 is localized to the calyx and is a testis-specific protein important for proper assembly of the calyx during spermatogenesis.
Between the head and the tail of spermatozoa is the connecting piece (neck region). It contains the two flagellar centrioles; one typical barrel-shaped proximal centriole and one atypical distal centriole, which is subject to compositional and structural remodelling during spermatogenesis Fishman EL et al. (2018). The proximal centriole is important for the connection to the sperm head and the nucleus. The distal centriole is connected to the tail, also known as the flagellum. The flagellum is about 10 times (55 μm) longer than the sperm head and divided into three structurally distinct parts: the mid piece, the principal piece, and the end piece. Common for all three parts is the microtubule-based axoneme that runs along the entire length of the flagellum. As for other motile cilia, the microtubules of the axoneme form a central pair surrounded by a ring of nine peripheral doublet microtubules linked by nexin and radial spokes, creating the characteristic "9+2" structure. The peripheral doublet microtubules are further supported by inner and outer dynein arms, which consist of motor proteins that help create the bending motion of the flagellum. In the mid piece and principal piece, the axoneme is surrounded by outer dense fibers that protect the cells from shear forces during movement. In the principal piece, two of the ODFs are replaced by the longitudinal columns of the fibrous sheat. The fibrous sheet protects and supports the axoneme, but also has a role in providing energy for flagellar motility though glycolysis. Sperm motility is mainly powered by ATP generated by a large number of mitochondria located in the mitochondrial sheath that coils around the axoneme of the mid piece (Dai C et al. (2021); Wang J et al. (2022); Dirami T et al. (2015)).
In vertebrates, a spetin-based ring-shaped structure called the annulus can be found between the mid piece and the principal piece. The annulus has been hypothesized to act as a diffusion barrier of proteins, guiding flagellar growth along the axoneme and the alignment of mitochondria in the mid piece. The end piece acks the fibrous sheet, and simply consist of the axoneme wrapped by the plasma membrane. In the most distal segment, the 9+2 doublet symmetry of the axoneme is lost, an the microtubules form singlets. The end piece is where the flagellum can grow and shrink, and it thus plays an important role in coordinating growth and intra‐flagellar transport. Examples of proteins localizing to the substructures of the neck and tail can be seen in Figure 4.
CATIP - Connecting piece
WDPCP - Flagellar centriole
RSPH1 - Mid piece
GAPDHS - Principal piece
DNAH2 - End piece
SEPTIN4 - Annulus
Figure 4. Examples showing the different sperm neck region and flagellar substructures and staining pattern CATIP is localized to the connecting piece and modulates actin polymerization. WDPCP is localized to the flagellar centriole and is a part of the CPLANE (ciliogenesis and planar polarity effectors) complex that is involved in the recruitment of interflagellar transport proteins to the basal body of cilia. RSPH1 is localized to the mid piece of the flagellum and is part of the radial spoke complex and is important for the motility of sperm. GAPDHS is localized to the principal piece of the flagellum and is a testis-specific enzyme that regulates the switch between different pathways for energy production in the sperm. DNAH2 is localized to the end piece of the flagellum and is a part of the inner dynein arm complex. SEPTIN4 is localized to the annulus where it is essential for its formation.
A selection of proteins suitable as markers for the sperm cell and its substructures can be found in Table 1.
Table 1. Selection of proteins suitable as markers for the sperm cell and its substructures.
Gene |
Description |
Substructure |
SPACA3
|
Sperm acrosome associated 3 |
Acrosome |
SEPTIN4
|
Septin 4 |
Annulus |
CYLC1
|
Cylicin 1 |
Calyx Perinuclear theca |
CYLC2
|
Cylicin 2 |
Calyx Perinuclear theca |
PMFBP1
|
Polyamine modulated factor 1 binding protein 1 |
Acrosome Connecting piece Flagellar centriole Mid piece Principal piece |
DNAH2
|
Dynein axonemal heavy chain 2 |
End piece Flagellar centriole |
SPESP1
|
Sperm equatorial segment protein 1 |
Acrosome Equatorial segment |
CEP131
|
Centrosomal protein 131 |
Flagellar centriole |
DNHD1
|
Dynein heavy chain domain 1 |
Annulus Mid piece |
CCIN
|
Calicin |
Calyx Perinuclear theca |
CABYR
|
Calcium binding tyrosine phosphorylation regulated |
Principal piece |
IQUB
|
IQ motif and ubiquitin domain containing |
End piece Mid piece Principal piece |
|
|
|
The function of the sperm cell and its substructures
The main function of the sperm cell is to carry the male genetic information to the oocyte during fertilization. Once the sperm cells are produced, they reside in an immature state in the testis. Upon ejaculation, sperm mature within the female reproductive tract. This maturation process is triggered by exposure to environmental cues, such as bicarbonate concentration, and leads to a more progressive flagellar beat and the capability to penetrate the egg vestments. The flagellum is used as the sperm cell's motor, rudder, and sensor, powered by the ATP produced by mitochondria in the mid piece. Flagellar movement results from the sliding of the axonemal microtubules under the action of dynein arms.
Multiple sensory proteins on the flagellar membrane, such as ion channels, detect signals and initiate a downstream signaling cascade that orchestrates the flagellar beat to modulate the swimming path (Alvarez L et al. (2014)).The sperm cell utilizes physical and chemical environmental cues, such as chemotaxis, haptotaxis, thermotaxis, or rheotaxis to navigate to the oocyte (Wachten D et al. (2017)). As sperm progress through the female reproductive tract and approach the egg, changes in the lipid and protein organization in a process known as capacitation, prepares the sperm to bind to the extracellular matrix that surrounds the egg (zona pellucida). The motility pattern of sperm also changes from being linear, to rapid and asymmetrical, in what is refered to as sperm hyperactiation. For successful fertilization, the sperm cell also need to go through the acrosome reaction, in which the acrosome membrane fuses with the plasma membrane of the spem head. This allows the acrosomal enzymes to degrade the extacellular matrix of the egg an exposes components necessary for binding to the underlying cell membrane of the egg. Membrane fusion then allows the sperm nucleus and the centrioles, but not the mitochondria to enter the egg, leading to the formation of a diploid zygote and initiation of embryonic development.
Infertility is a widespread condition that affect millions of couples around the globe (Cannarella R et al. (2020)). In half of the cases, male infertility causes or contributes to the inablility to achieve conception. This may result from defects during spermatogenesis, defects in sperm functionality in terms of reaching and fusing with the egg, or from factors that affect embryonal growth and development. While it was originally thought that the male contribution to fertilization was resticted to delivery of sperm DNA, recent years of studies have shown that proteins as well as RNA originating from the sperm cell are important for various aspects of fertilization and embryogenesis. In as much as 70% of the cases, the molecular targets of male infertility remains elusive, pointing towards a need for further studies of mechanisms and pathways underpinning sperm structure and function, including the detailed spatial architecture of proteins that can be found in sperm.
Gene Ontology (GO)-based functional enrichment analysis of genes encoding sperm proteins shows an enrichment of terms that are well in line with the known functions of the sperm cell. The most highly enriched terms for the GO domain Biological Process are related to sperm motility, assembly and organization of the cilium, and processes related to development (Figure 5a). Enrichment analysis of the GO domain Molecular Function reveal enrichment of terms describing binding to microtubules and motor proteins, as well as motor activity (Figure 5b).
Figure 5a. Gene Ontology-based enrichment analysis for the sperm 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 sperm 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.
Sperm proteins with multiple locations
In the subcellular resource, approximately 76% (n=493) of the proteins that localize to the sperm cell and its substructures also localize to other cell compartments in other cell types (Figure 6). The network plot shows that the most common locations shared with the sperm cell are the mitochondria, cytosol, and nucleoplasm. Dual localizations with the mitochondria are overrepresented, while dual localizations with the plasma membrane are underrepresented.
Figure 6. Interactive network plot of sperm cell proteins with multiple localizations. The numbers in the connecting nodes show the proteins that are localized to the sperm cell and its substructures, and to one or more additional locations in other cell types. Only connecting nodes containing at least 1.0% of the proteins in the sperm 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.
Expression levels of sperm proteins in tissue
Transcriptome analysis and classification of genes into tissue distribution categories (Figure 7) shows that the genes encoding proteins localizing to the sperm cell and its substructures are more likely to be detected in a single or some tissues, but less likely to be ubiquitously expressed compared to all genes presented within the subcellular resource. Thus, these genes show a more restricted pattern of tissue expression compared to other genes, likely reflecting the specific function of the sperm cell and the many sperm-specific proteins.
Figure 7. Bar plot showing the percentage of genes in different tissue distribution categories for sperm 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
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
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
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
Norreen-Thorsen M et al., A human adipose tissue cell-type transcriptome atlas. Cell Rep. (2022)
PubMed: 35830816 DOI: 10.1016/j.celrep.2022.111046
Öling S et al., A human stomach cell type transcriptome atlas. BMC Biol. (2024)
PubMed: 38355543 DOI: 10.1186/s12915-024-01812-5
Siletti K et al., Transcriptomic diversity of cell types across the adult human brain. Science. (2023)
PubMed: 37824663 DOI: 10.1126/science.add7046
Uhlén M et al., The human secretome. Sci Signal. (2019)
PubMed: 31772123 DOI: 10.1126/scisignal.aaz0274
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
Uhlen M et al., A pathology atlas of the human cancer transcriptome. Science. (2017)
PubMed: 28818916 DOI: 10.1126/science.aan2507
Li Y et al., Proteogenomic data and resources for pan-cancer analysis. Cancer Cell. (2023)
PubMed: 37582339 DOI: 10.1016/j.ccell.2023.06.009
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
Sakalihasan N et al., Abdominal aortic aneurysms. Nat Rev Dis Primers. (2018)
PubMed: 30337540 DOI: 10.1038/s41572-018-0030-7
Krafcik BM et al., Changes in global mortality from aortic aneurysm. J Vasc Surg. (2024)
PubMed: 38408686 DOI: 10.1016/j.jvs.2024.02.025
Hultgren R et al., A Majority of Admitted Patients With Ruptured Abdominal Aortic Aneurysm Undergo and Survive Corrective Treatment: A Population-Based Retrospective Cohort Study. World J Surg. (2016)
PubMed: 27549597 DOI: 10.1007/s00268-016-3705-9
Shanmuganathan G et al., Diabetes and Abdominal Aortic Aneurysm: Is the Protective Effect on AAA Due to Antidiabetic Medications Alone, Due to the Disease Alone, or Both? Arch Intern Med Res. (2024)
PubMed: 38846325 DOI: 10.26502/aimr.0169
Bugiardini R et al., Traditional risk factors and premature acute coronary syndromes in South Eastern Europe: a multinational cohort study. Lancet Reg Health Eur. (2024)
PubMed: 38476741 DOI: 10.1016/j.lanepe.2023.100824
Timmis A et al., Global epidemiology of acute coronary syndromes. Nat Rev Cardiol. (2023)
PubMed: 37231077 DOI: 10.1038/s41569-023-00884-0
Shimony S et al., Acute myeloid leukemia: 2023 update on diagnosis, risk-stratification, and management. Am J Hematol. (2023)
PubMed: 36594187 DOI: 10.1002/ajh.26822
Pelcovits A et al., Acute Myeloid Leukemia: A Review. R I Med J (2013). (2020)
PubMed: 32236160
Yi M et al., The global burden and attributable risk factor analysis of acute myeloid leukemia in 195 countries and territories from 1990 to 2017: estimates based on the global burden of disease study 2017. J Hematol Oncol. (2020)
PubMed: 32513227 DOI: 10.1186/s13045-020-00908-z
Søgaard KK et al., 30-year mortality after venous thromboembolism: a population-based cohort study. Circulation. (2014)
PubMed: 24970783 DOI: 10.1161/CIRCULATIONAHA.114.009107
O'Shea RS et al., Alcoholic liver disease. Am J Gastroenterol. (2010)
PubMed: 19904248 DOI: 10.1038/ajg.2009.593
Gao B et al., Alcoholic liver disease: pathogenesis and new therapeutic targets. Gastroenterology. (2011)
PubMed: 21920463 DOI: 10.1053/j.gastro.2011.09.002
Stickel F et al., Pathophysiology and Management of Alcoholic Liver Disease: Update 2016. Gut Liver. (2017)
PubMed: 28274107 DOI: 10.5009/gnl16477
Sharma P et al., Clinical presentation of alcoholic liver disease and non-alcoholic fatty liver disease: spectrum and diagnosis. Transl Gastroenterol Hepatol. (2020)
PubMed: 32258523 DOI: 10.21037/tgh.2019.10.02
Mathurin P et al., Corticosteroids improve short-term survival in patients with severe alcoholic hepatitis: meta-analysis of individual patient data. Gut. (2011)
PubMed: 20940288 DOI: 10.1136/gut.2010.224097
Sattar SBA et al., Bacterial Gastroenteritis StatPearls Publishing. (2024)
PubMed: 30020667
Nierenberg AA et al., Diagnosis and Treatment of Bipolar Disorder: A Review. JAMA. (2023)
PubMed: 37815563 DOI: 10.1001/jama.2023.18588
Lane NM et al., Bipolar disorder: Diagnosis, treatment and future directions. J R Coll Physicians Edinb. (2023)
PubMed: 37649414 DOI: 10.1177/14782715231197577
Wang J et al., Progression from ductal carcinoma in situ to invasive breast cancer: molecular features and clinical significance. Signal Transduct Target Ther. (2024)
PubMed: 38570490 DOI: 10.1038/s41392-024-01779-3
Tomlinson-Hansen SE et al., Breast Ductal Carcinoma in Situ StatPearls Publishing. (2024)
PubMed: 33620843
Chowdhury AB et al., Liver biopsy for assessment of chronic liver diseases: a synopsis. Clin Exp Med. (2023)
PubMed: 35192111 DOI: 10.1007/s10238-022-00799-z
Ginès P et al., Population screening for liver fibrosis: Toward early diagnosis and intervention for chronic liver diseases. Hepatology. (2022)
PubMed: 34537988 DOI: 10.1002/hep.32163
Heyens LJM et al., Liver Fibrosis in Non-alcoholic Fatty Liver Disease: From Liver Biopsy to Non-invasive Biomarkers in Diagnosis and Treatment. Front Med (Lausanne). (2021)
PubMed: 33937277 DOI: 10.3389/fmed.2021.615978
Asrani SK et al., Burden of liver diseases in the world. J Hepatol. (2019)
PubMed: 30266282 DOI: 10.1016/j.jhep.2018.09.014
Mukkamalla SKR et al., Chronic Lymphocytic Leukemia StatPearls Publishing. (2024)
PubMed: 29261864
Kronmal RA et al., Risk factors for the progression of coronary artery calcification in asymptomatic subjects: results from the Multi-Ethnic Study of Atherosclerosis (MESA). Circulation. (2007)
PubMed: 17502571 DOI: 10.1161/CIRCULATIONAHA.106.674143
Roth GA et al., Global Burden of Cardiovascular Diseases and Risk Factors, 1990-2019: Update From the GBD 2019 Study. J Am Coll Cardiol. (2020)
PubMed: 33309175 DOI: 10.1016/j.jacc.2020.11.010
Begum F et al., Insight into the Tropism of Dengue Virus in Humans. Viruses. (2019)
PubMed: 31835302 DOI: 10.3390/v11121136
Blackley S et al., Primary human splenic macrophages, but not T or B cells, are the principal target cells for dengue virus infection in vitro. J Virol. (2007)
PubMed: 17928355 DOI: 10.1128/JVI.01568-07
Padala SA et al., Diffuse Large B-Cell Lymphoma StatPearls Publishing. (2024)
PubMed: 32491728
Ramakrishnan K et al., Diagnosis and management of acute pyelonephritis in adults. Am Fam Physician. (2005)
PubMed: 15768623
Makker V et al., Endometrial cancer. Nat Rev Dis Primers. (2021)
PubMed: 34887451 DOI: 10.1038/s41572-021-00324-8
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
Ostrom QT et al., The epidemiology of glioma in adults: a "state of the science" review. Neuro Oncol. (2014)
PubMed: 24842956 DOI: 10.1093/neuonc/nou087
Goodenberger ML et al., Genetics of adult glioma. Cancer Genet. (2012)
PubMed: 23238284 DOI: 10.1016/j.cancergen.2012.10.009
Llovet JM et al., Hepatocellular carcinoma. Nat Rev Dis Primers. (2021)
PubMed: 33479224 DOI: 10.1038/s41572-020-00240-3
Matsushita H et al., Alcohol and hepatocellular carcinoma. BMJ Open Gastroenterol. (2019)
PubMed: 31139422 DOI: 10.1136/bmjgast-2018-000260
Singal AG et al., HCC surveillance improves early detection, curative treatment receipt, and survival in patients with cirrhosis: A meta-analysis. J Hepatol. (2022)
PubMed: 35139400 DOI: 10.1016/j.jhep.2022.01.023
Tzartzeva K et al., Surveillance Imaging and Alpha Fetoprotein for Early Detection of Hepatocellular Carcinoma in Patients With Cirrhosis: A Meta-analysis. Gastroenterology. (2018)
PubMed: 29425931 DOI: 10.1053/j.gastro.2018.01.064
Bruix J et al., Management of hepatocellular carcinoma: an update. Hepatology. (2011)
PubMed: 21374666 DOI: 10.1002/hep.24199
El-Khoueiry AB et al., Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet. (2017)
PubMed: 28434648 DOI: 10.1016/S0140-6736(17)31046-2
Calderon-Martinez E et al., Prognostic Scores and Survival Rates by Etiology of Hepatocellular Carcinoma: A Review. J Clin Med Res. (2023)
PubMed: 37187717 DOI: 10.14740/jocmr4902
Royle CM et al., HIV-1 and HIV-2 differentially mature plasmacytoid dendritic cells into IFN-producing cells or APCs. J Immunol. (2014)
PubMed: 25156368 DOI: 10.4049/jimmunol.1400860
Gaitonde DY et al., Influenza: Diagnosis and Treatment. Am Fam Physician. (2019)
PubMed: 31845781
Wu NH et al., The differentiated airway epithelium infected by influenza viruses maintains the barrier function despite a dramatic loss of ciliated cells. Sci Rep. (2016)
PubMed: 28004801 DOI: 10.1038/srep39668
Nasim F et al., Lung Cancer. Med Clin North Am. (2019)
PubMed: 30955514 DOI: 10.1016/j.mcna.2018.12.006
Buck E et al., Malaria StatPearls Publishing. (2024)
PubMed: 31869175
Milner DA., Malaria Pathogenesis. Cold Spring Harb Perspect Med. (2018)
PubMed: 28533315 DOI: 10.1101/cshperspect.a025569
Sanyal AJ et al., Cardiovascular disease in patients with metabolic dysfunction-associated steatohepatitis compared with metabolic dysfunction-associated steatotic liver disease and other liver diseases: A systematic review. Am Heart J Plus. (2024)
PubMed: 38623572 DOI: 10.1016/j.ahjo.2024.100386
Marjot T et al., Nonalcoholic Fatty Liver Disease in Adults: Current Concepts in Etiology, Outcomes, and Management. Endocr Rev. (2020)
PubMed: 31629366 DOI: 10.1210/endrev/bnz009
Chan WK et al., Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): A State-of-the-Art Review. J Obes Metab Syndr. (2023)
PubMed: 37700494 DOI: 10.7570/jomes23052
Hashimoto E et al., Characteristics and diagnosis of NAFLD/NASH. J Gastroenterol Hepatol. (2013)
PubMed: 24251707 DOI: 10.1111/jgh.12271
Heistein JB et al., Malignant Melanoma StatPearls Publishing. (2024)
PubMed: 29262210
Rastrelli M et al., Melanoma: epidemiology, risk factors, pathogenesis, diagnosis and classification. In Vivo. (2014)
PubMed: 25398793
Alruwaili AA et al., Meningioma StatPearls Publishing. (2024)
PubMed: 32809373
Saklayen MG., The Global Epidemic of the Metabolic Syndrome. Curr Hypertens Rep. (2018)
PubMed: 29480368 DOI: 10.1007/s11906-018-0812-z
Eckel RH et al., The metabolic syndrome. Lancet. (2010)
PubMed: 20109902 DOI: 10.1016/S0140-6736(09)61794-3
Grundy SM et al., Diagnosis and management of the metabolic syndrome: an American Heart Association/National Heart, Lung, and Blood Institute Scientific Statement. Circulation. (2005)
PubMed: 16157765 DOI: 10.1161/CIRCULATIONAHA.105.169404
Lim Y et al., Obesity and Comorbid Conditions StatPearls Publishing. (2024)
PubMed: 34662049
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
Kotol D et al., Absolute Quantification of Pan-Cancer Plasma Proteomes Reveals Unique Signature in Multiple Myeloma. Cancers (Basel). (2023)
PubMed: 37835457 DOI: 10.3390/cancers15194764
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
Ritchie ME et al., limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. (2015)
PubMed: 25605792 DOI: 10.1093/nar/gkv007
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
Haki M et al., Review of multiple sclerosis: Epidemiology, etiology, pathophysiology, and treatment. Medicine (Baltimore). (2024)
PubMed: 38394496 DOI: 10.1097/MD.0000000000037297
Doshi A et al., Multiple sclerosis, a treatable disease. Clin Med (Lond). (2016)
PubMed: 27956442 DOI: 10.7861/clinmedicine.16-6-s53
Dobson R et al., Multiple sclerosis - a review. Eur J Neurol. (2019)
PubMed: 30300457 DOI: 10.1111/ene.13819
Lundberg IE et al., Classification of myositis. Nat Rev Rheumatol. (2018)
PubMed: 29651121 DOI: 10.1038/nrrheum.2018.41
Ashton C et al., Idiopathic inflammatory myopathies: a review. Intern Med J. (2021)
PubMed: 34155760 DOI: 10.1111/imj.15358
Panuganti KK et al., Obesity StatPearls Publishing. (2024)
PubMed: 29083734
Blüher M., Obesity: global epidemiology and pathogenesis. Nat Rev Endocrinol. (2019)
PubMed: 30814686 DOI: 10.1038/s41574-019-0176-8
Brod M et al., Development of the Weight-Related Sign and Symptom Measure. J Patient Rep Outcomes. (2017)
PubMed: 29757304 DOI: 10.1186/s41687-018-0042-9
Ghesmaty Sangachin M et al., Use of various obesity measurement and classification methods in occupational safety and health research: a systematic review of the literature. BMC Obes. (2018)
PubMed: 30410773 DOI: 10.1186/s40608-018-0205-5
Arora T et al., Epithelial Ovarian Cancer StatPearls Publishing. (2024)
PubMed: 33620837
Zhang R et al., Molecular Biomarkers for the Early Detection of Ovarian Cancer. Int J Mol Sci. (2022)
PubMed: 36233339 DOI: 10.3390/ijms231912041
Penny SM., Ovarian Cancer: An Overview. Radiol Technol. (2020)
PubMed: 32606233
Ilic M et al., Epidemiology of pancreatic cancer. World J Gastroenterol. (2016)
PubMed: 27956793 DOI: 10.3748/wjg.v22.i44.9694
Leblond P et al., Toward Improved Diagnosis Accuracy and Treatment of Children, Adolescents, and Young Adults With Ependymoma: The International SIOP Ependymoma II Protocol. Front Neurol. (2022)
PubMed: 35720069 DOI: 10.3389/fneur.2022.887544
Tauziède-Espariat A et al., Pediatric meningiomas: A literature review and diagnostic update. Neurooncol Adv. (2023)
PubMed: 37287580 DOI: 10.1093/noajnl/vdac165
Campen CJ et al., Optic Pathway Gliomas in Neurofibromatosis Type 1. J Child Neurol. (2018)
PubMed: 29246098 DOI: 10.1177/0883073817739509
Oronsky B et al., Nothing But NET: A Review of Neuroendocrine Tumors and Carcinomas. Neoplasia. (2017)
PubMed: 29091800 DOI: 10.1016/j.neo.2017.09.002
Smolen JS et al., Rheumatoid arthritis. Lancet. (2016)
PubMed: 27156434 DOI: 10.1016/S0140-6736(16)30173-8
Radu AF et al., Management of Rheumatoid Arthritis: An Overview. Cells. (2021)
PubMed: 34831081 DOI: 10.3390/cells10112857
Patel KR et al., Schizophrenia: overview and treatment options. P T. (2014)
PubMed: 25210417
Carsons SE et al., Sjogren Syndrome StatPearls Publishing. (2024)
PubMed: 28613703
Jonsson R et al., Current concepts on Sjögren's syndrome - classification criteria and biomarkers. Eur J Oral Sci. (2018)
PubMed: 30178554 DOI: 10.1111/eos.12536
Gonzáles-Yovera JG et al., Diagnosis and management of small bowel neuroendocrine tumors: A state-of-the-art. World J Methodol. (2022)
PubMed: 36186753 DOI: 10.5662/wjm.v12.i5.381
Tong SY et al., Staphylococcus aureus infections: epidemiology, pathophysiology, clinical manifestations, and management. Clin Microbiol Rev. (2015)
PubMed: 26016486 DOI: 10.1128/CMR.00134-14
Abraham L et al., Bacteremia: Contemporary Management. Mo Med. (2020)
PubMed: 32848271
Bruun T et al., Risk Factors and Predictors of Mortality in Streptococcal Necrotizing Soft-tissue Infections: A Multicenter Prospective Study. Clin Infect Dis. (2021)
PubMed: 31923305 DOI: 10.1093/cid/ciaa027
Hua C et al., Necrotising soft-tissue infections. Lancet Infect Dis. (2023)
PubMed: 36252579 DOI: 10.1016/S1473-3099(22)00583-7
Peetermans M et al., Necrotizing skin and soft-tissue infections in the intensive care unit. Clin Microbiol Infect. (2020)
PubMed: 31284035 DOI: 10.1016/j.cmi.2019.06.031
Madsen MB et al., Patient's characteristics and outcomes in necrotising soft-tissue infections: results from a Scandinavian, multicentre, prospective cohort study. Intensive Care Med. (2019)
PubMed: 31440795 DOI: 10.1007/s00134-019-05730-x
Yu H et al., Clinical and Immunological Biomarkers for Systemic Lupus Erythematosus. Biomolecules. (2021)
PubMed: 34206696 DOI: 10.3390/biom11070928
Siegel CH et al., Systemic Lupus Erythematosus: A Review. JAMA. (2024)
PubMed: 38587826 DOI: 10.1001/jama.2024.2315
Lazar S et al., Systemic Lupus Erythematosus: New Diagnostic and Therapeutic Approaches. Annu Rev Med. (2023)
PubMed: 35804480 DOI: 10.1146/annurev-med-043021-032611
Volkmann ER et al., Systemic sclerosis. Lancet. (2023)
PubMed: 36442487 DOI: 10.1016/S0140-6736(22)01692-0
Peoples C et al., Gender differences in systemic sclerosis: relationship to clinical features, serologic status and outcomes. J Scleroderma Relat Disord. (2016)
PubMed: 29242839 DOI: 10.5301/jsrd.5000209
Ahlqvist E et al., Novel subgroups of adult-onset diabetes and their association with outcomes: a data-driven cluster analysis of six variables. Lancet Diabetes Endocrinol. (2018)
PubMed: 29503172 DOI: 10.1016/S2213-8587(18)30051-2
American Diabetes Association., 2. Classification and Diagnosis of Diabetes: . Diabetes Care. (2021)
PubMed: 33298413 DOI: 10.2337/dc21-S002
Zheng Y et al., Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nat Rev Endocrinol. (2018)
PubMed: 29219149 DOI: 10.1038/nrendo.2017.151
Khan MAB et al., Epidemiology of Type 2 Diabetes - Global Burden of Disease and Forecasted Trends. J Epidemiol Glob Health. (2020)
PubMed: 32175717 DOI: 10.2991/jegh.k.191028.001
Sharma A et al., Chronic Liver Disease StatPearls Publishing. (2024)
PubMed: 32119484
Nelson NP et al., Epidemiology of Hepatitis B Virus Infection and Impact of Vaccination on Disease. Clin Liver Dis. (2016)
PubMed: 27742003 DOI: 10.1016/j.cld.2016.06.006
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
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
Smolders J et al., Tissue-resident memory T cells populate the human brain. Nat Commun. (2018)
PubMed: 30389931 DOI: 10.1038/s41467-018-07053-9
Jain RW et al., B cells in central nervous system disease: diversity, locations and pathophysiology. Nat Rev Immunol. (2022)
PubMed: 34903877 DOI: 10.1038/s41577-021-00652-6
Waller R et al., Iba-1-/CD68+ microglia are a prominent feature of age-associated deep subcortical white matter lesions. PLoS One. (2019)
PubMed: 30682074 DOI: 10.1371/journal.pone.0210888
Buchanan J et al., Oligodendrocyte precursor cells ingest axons in the mouse neocortex. Proc Natl Acad Sci U S A. (2022)
PubMed: 36417438 DOI: 10.1073/pnas.2202580119
Fang M et al., The committed oligodendrocyte precursor cell, a newly-defined intermediate progenitor cell type in oligodendroglial lineage. Glia. (2023)
PubMed: 37278537 DOI: 10.1002/glia.24426
Duan L et al., Fibroblasts: New players in the central nervous system? Fundam Res. (2024)
PubMed: 38933505 DOI: 10.1016/j.fmre.2023.01.014
Lendahl U et al., Identification, discrimination and heterogeneity of fibroblasts. Nat Commun. (2022)
PubMed: 35701396 DOI: 10.1038/s41467-022-30633-9
Wong FK et al., Serotonergic regulation of bipolar cell survival in the developing cerebral cortex. Cell Rep. (2022)
PubMed: 35793629 DOI: 10.1016/j.celrep.2022.111037
Lv X et al., TBR2 coordinates neurogenesis expansion and precise microcircuit organization via Protocadherin 19 in the mammalian cortex. Nat Commun. (2019)
PubMed: 31477701 DOI: 10.1038/s41467-019-11854-x
Robinson JL et al., An atlas of human metabolism. Sci Signal. (2020)
PubMed: 32209698 DOI: 10.1126/scisignal.aaz1482
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
Cheng J et al., Accurate proteome-wide missense variant effect prediction with AlphaMissense. Science. (2023)
PubMed: 37733863 DOI: 10.1126/science.adg7492
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
Li F et al., GotEnzymes: an extensive database of enzyme parameter predictions. Nucleic Acids Res. (2023)
PubMed: 36169223 DOI: 10.1093/nar/gkac831
Bar-Peled L et al., Principles and functions of metabolic compartmentalization. Nat Metab. (2022)
PubMed: 36266543 DOI: 10.1038/s42255-022-00645-2
Song J et al., Assembling the mitochondrial ATP synthase. Proc Natl Acad Sci U S A. (2018)
PubMed: 29514954 DOI: 10.1073/pnas.1801697115
Maxfield FR et al., Cholesterol, the central lipid of mammalian cells. Curr Opin Cell Biol. (2010)
PubMed: 20627678 DOI: 10.1016/j.ceb.2010.05.004
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
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
Mill P et al., Primary cilia as dynamic and diverse signalling hubs in development and disease. Nat Rev Genet. (2023)
PubMed: 37072495 DOI: 10.1038/s41576-023-00587-9
Macarelli V et al., Regulation of the length of neuronal primary cilia and its potential effects on signalling. Trends Cell Biol. (2023)
PubMed: 37302961 DOI: 10.1016/j.tcb.2023.05.005
Satir P et al., The primary cilium at a glance. J Cell Sci. (2010)
PubMed: 20144997 DOI: 10.1242/jcs.050377
Taschner M et al., The Intraflagellar Transport Machinery. Cold Spring Harb Perspect Biol. (2016)
PubMed: 27352625 DOI: 10.1101/cshperspect.a028092
Nachury MV et al., Establishing and regulating the composition of cilia for signal transduction. Nat Rev Mol Cell Biol. (2019)
PubMed: 30948801 DOI: 10.1038/s41580-019-0116-4
Reiter JF et al., Genes and molecular pathways underpinning ciliopathies. Nat Rev Mol Cell Biol. (2017)
PubMed: 28698599 DOI: 10.1038/nrm.2017.60
Anvarian Z et al., Cellular signalling by primary cilia in development, organ function and disease. Nat Rev Nephrol. (2019)
PubMed: 30733609 DOI: 10.1038/s41581-019-0116-9
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
Uhlén M et al., Tissue-based map of the human proteome. Science (2015)
PubMed: 25613900 DOI: 10.1126/science.1260419
Fagerberg L et al., Prediction of the human membrane proteome. Proteomics. (2010)
PubMed: 20175080 DOI: 10.1002/pmic.200900258
Schneider S et al., Cylicins are a structural component of the sperm calyx being indispensable for male fertility in mice and human. Elife. (2023)
PubMed: 38013430 DOI: 10.7554/eLife.86100
Balhorn R., The protamine family of sperm nuclear proteins. Genome Biol. (2007)
PubMed: 17903313 DOI: 10.1186/gb-2007-8-9-227
Khawar MB et al., Mechanism of Acrosome Biogenesis in Mammals. Front Cell Dev Biol. (2019)
PubMed: 31620437 DOI: 10.3389/fcell.2019.00195
Fishman EL et al., A novel atypical sperm centriole is functional during human fertilization. Nat Commun. (2018)
PubMed: 29880810 DOI: 10.1038/s41467-018-04678-8
Dai C et al., Advances in sperm analysis: techniques, discoveries and applications. Nat Rev Urol. (2021)
PubMed: 34075227 DOI: 10.1038/s41585-021-00472-2
Wang J et al., Clinical detection, diagnosis and treatment of morphological abnormalities of sperm flagella: A review of literature. Front Genet. (2022)
PubMed: 36425067 DOI: 10.3389/fgene.2022.1034951
Dirami T et al., Assessment of the frequency of sperm annulus defects in a large cohort of patients presenting asthenozoospermia. Basic Clin Androl. (2015)
PubMed: 26576287 DOI: 10.1186/s12610-015-0026-z
Alvarez L et al., The computational sperm cell. Trends Cell Biol. (2014)
PubMed: 24342435 DOI: 10.1016/j.tcb.2013.10.004
Wachten D et al., Sperm Sensory Signaling. Cold Spring Harb Perspect Biol. (2017)
PubMed: 28062561 DOI: 10.1101/cshperspect.a028225
Cannarella R et al., Molecular Biology of Spermatogenesis: Novel Targets of Apparently Idiopathic Male Infertility. Int J Mol Sci. (2020)
PubMed: 32138324 DOI: 10.3390/ijms21051728