RAD21

Last updated
RAD21
Available structures
PDB Ortholog search: PDBe RCSB
Identifiers
Aliases RAD21 , CDLS4, HR21, HMCD1, NXP1, SCC1, hHR21, RAD21 cohesin complex component, MGS
External IDs OMIM: 606462 MGI: 108016 HomoloGene: 38161 GeneCards: RAD21
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_006265

NM_009009

RefSeq (protein)

NP_006256

NP_033035

Location (UCSC) Chr 8: 116.85 – 116.87 Mb Chr 15: 51.83 – 51.86 Mb
PubMed search [3] [4]
Wikidata
View/Edit Human View/Edit Mouse

Double-strand-break repair protein rad21 homolog is a protein that in humans is encoded by the RAD21 gene. [5] [6] RAD21 (also known as Mcd1, Scc1, KIAA0078, NXP1, HR21), an essential gene, encodes a DNA double-strand break (DSB) repair protein that is evolutionarily conserved in all eukaryotes from budding yeast to humans.  RAD21 protein is a structural component of the highly conserved cohesin complex consisting of RAD21, SMC1A, SMC3, and SCC3 [ STAG1 (SA1) and STAG2 (SA2) in multicellular organisms] proteins, involved in sister chromatid cohesion.

Contents

Discovery

rad21 was first cloned by Birkenbihl and Subramani in 1992 [7] by complementing the radiation sensitivity of the rad21-45 mutant fission yeast, Schizosaccharomyces pombe , and the murine and human homologs of S. pombe rad21 were cloned by McKay, Troelstra, van der Spek, Kanaar, Smit, Hagemeijer, Bootsma and Hoeijmakers. [8]   The human RAD21 (hRAD21) gene is located on the long (q) arm of chromosome 8 at position 24.11 (8q24.11). [8] [9]   In 1997, RAD21 was independently discovered by two groups to be a major component of the chromosomal cohesin complex, [10] [11] and its dissolution by the cysteine protease Separase at the metaphase to anaphase transition results in the separation of sister chromatids and chromosomal segregation. [12]

Structure

RAD21, belongs to a superfamily of eukaryotic and prokaryotic proteins called a-Kleisins, [13] is a nuclear phospho-protein, ranges in size from 278aa in the house lizard ( Gekko Japonicus ) to 746aa in the killer whale ( Orcinus Orca ), with a median length of 631aa in most vertebrate species including humans.  RAD21 proteins are most conserved at the N-terminus (NT) and C-terminus (CT), which bind to SMC3 and SMC1, respectively. The STAG domain in the middle of RAD21, which binds to SCC3 (SA1/SA2), is also conserved (Figure 1). These proteins have nuclear localization signals, an acidic-basic stretch and an acidic stretch (Figure 1), which is consistent with a chromatin-binding role. RAD21 is cleaved by several proteases including Separase [12] [14] [15] and Calcium-dependent cysteine endopeptidase Calpain-1 [16] during mitosis and Caspases during apoptosis. [17] [18]

Figure 1 Characteristics on human RAD21. RAD21 has three binding domains that interact with corresponding protein: SMC3 (1-103aa), STAG1/2 (362-403aa) and SMC1A (558-628aa); a LPE motif (255-257aa): required for rapid and specific cleavage of RAD21 by Separase; two bipartite nuclear localization signals (NLS) (317-399aa and 384-407aa) predicted by cNLS Mapper; one alternating acidic-basic residues stretch (524-533aa); one acidic residues stretch (534-543aa); four cleavage sites: two Separase cleavage sites (ExxR), one Calpain-1 cleavage site (LLL) and one Caspase-3/7 site (DxxD). The numbers indicate the location of amino acid residue on human RAD21.  The arrow shows the site where it is cleaved. Characteristics on human RAD21. .tif
Figure 1 Characteristics on human RAD21. RAD21 has three binding domains that interact with corresponding protein: SMC3 (1–103aa), STAG1/2 (362–403aa) and SMC1A (558–628aa); a LPE motif (255-257aa): required for rapid and specific cleavage of RAD21 by Separase; two bipartite nuclear localization signals (NLS) (317-399aa and 384-407aa) predicted by cNLS Mapper; one alternating acidic-basic residues stretch (524-533aa); one acidic residues stretch (534-543aa); four cleavage sites: two Separase cleavage sites (ExxR), one Calpain-1 cleavage site (LLL) and one Caspase-3/7 site (DxxD). The numbers indicate the location of amino acid residue on human RAD21.  The arrow shows the site where it is cleaved.

Interactions

RAD21 binds to the V-shaped SMC1 and SMC3 heterodimer, forming a tripartite ring-like structure, [20] and then recruits SCC3 (SA1/SA2). The 4 element-complex is called the cohesin complex (Figure 2).  Currently, there are two major competing models of sister chromatid cohesion (Figure 2B).  The first one is the one-ring embrace model, [21] and the second one is the dimeric handcuff-model. [22] [23]  The one-ring embrace model posits that a single cohesin ring traps two sister chromatids inside, while the two-ring handcuff model proposes trapping of each chromatid individually.  According to the handcuff model, each ring has one set of RAD21, SMC1, and SMC3 molecules. The handcuff is established when two RAD21 molecules move into anti-parallel orientation that is enforced by either SA1 or SA2. [22]

Figure 2 Cohesin complex and models. A. Cohesin is composed of four core structural subunits: RAD21, SMC1, SMC3, and a SA protein (SA1 or SA2).  PDS5, WAPL, and Sororin are cohesin-associate proteins. Sororin has not been found in yeast. B. One-ring model. C. Handcuff model.  Figure adapted with modifications from Zhang and Pati. Figure 2 Cohesin complex and models. .tif
Figure 2 Cohesin complex and models. A. Cohesin is composed of four core structural subunits: RAD21, SMC1, SMC3, and a SA protein (SA1 or SA2).  PDS5, WAPL, and Sororin are cohesin-associate proteins. Sororin has not been found in yeast. B. One-ring model. C. Handcuff model.  Figure adapted with modifications from Zhang and Pati.

The N-terminal domain of RAD21 contains two α-helices that forms a three helical bundle with the coiled coil of SMC3. [20] The central region of RAD21 is thought to be largely unstructured but contains several binding sites for regulators of cohesin. This includes a binding site for SA1 or SA2, [27] recognition motifs for separase, caspase, and calpain to cleave, [12] [16] [17] [18] as well as a region that is competitively bound by PDS5A, PDS5B or NIPBL. [28] [29] [30]   The C-terminal domain of RAD21 forms a winged helix that binds two β-sheets in the Smc1 head domain. [31]

WAPL releases cohesin from DNA by opening the SMC3-RAD21 interface thereby allowing DNA to pass out of the ring. [32]   Opening of this interface is regulated by ATP-binding by the SMC subunits. This causes the ATPase head domains to dimerise and deforms the coiled coil of SMC3 therefore disrupting the binding of RAD21 to the coiled coil. [33]

Figure 3 Functional classification of RAD21 interactors. Fig output by Cytoscape with the data retrieved from Panigrahi et al. 2012. Network nodes represent proteins.  Edges represent protein-protein associations, clustered in different cellular processes. Figure 3 Functional classification of RAD21 interactors..tif
Figure 3 Functional classification of RAD21 interactors. Fig output by Cytoscape with the data retrieved from Panigrahi et al. 2012. Network nodes represent proteins.  Edges represent protein-protein associations, clustered in different cellular processes.

A total of 285 RAD21-interactants have been reported [34] that function in wide range of cellular processes, including mitosis, regulation of apoptosis, chromosome dynamics, chromosomal cohesion, replication, transcription regulation, RNA processing, DNA damage response, protein modification and degradation, and cytoskeleton and cell motility (Figure 3). [35]

Function

Figure 4 RAD21 Functions in various cellular processes. RAD21 forms cohesin complex with SMC1, SMC3 and STAG1/2 to function in various normal cellular processes (shown in blue). The canonical role of Rad21 is sister chromatid cohesion and separation. Other roles include DNA damage repair, transcription regulation, DNA replication, and centrosome biogenesis, etc. Diseases rise when mutations in RAD21 disrupt its function (in green). Caspase-cleaved Rad21 fragment promotes apoptosis (in purple). REC8 and RAD21L are paralogs of RAD21 in vertebrate, which function specifically to meiosis (in brown). Figure 4 RAD21 Functions in various cellular processes. .tif
Figure 4 RAD21 Functions in various cellular processes. RAD21 forms cohesin complex with SMC1, SMC3 and STAG1/2 to function in various normal cellular processes (shown in blue). The canonical role of Rad21 is sister chromatid cohesion and separation. Other roles include DNA damage repair, transcription regulation, DNA replication, and centrosome biogenesis, etc. Diseases rise when mutations in RAD21 disrupt its function (in green). Caspase-cleaved Rad21 fragment promotes apoptosis (in purple). REC8 and RAD21L are paralogs of RAD21 in vertebrate, which function specifically to meiosis (in brown).

RAD21 plays multiple physiological roles in diverse cellular functions (Figure 4).  As a subunit of the cohesin complex, RAD21 is involved in sister chromatid cohesion from the time of DNA replication in S phase to their segregation in mitosis, a function that is evolutionarily conserved and essential for proper chromosome segregation, chromosomal architecture, post-replicative DNA repair, and the prevention of inappropriate recombination between repetitive regions. [14] [26] RAD21 may also play a role in spindle pole assembly during mitosis [36] and progression of apoptosis. [17] [18] In interphase, cohesin may function in the control of gene expression by binding to numerous sites within the genome. As a structural component of the cohesin complex, RAD21 also contributes to various chromatin-associated functions, including DNA replication, [37] [38] [39] [40] [41] DNA damage response (DDR), [42] [43] [44] [45] [46] [47] [48] [49] [50] and most importantly, transcriptional regulation. [51] [52] [53] [54] [55] [56] [57] [58]   Numerous recent functional and genomic studies have implicated chromosomal cohesin proteins as critical regulators of hematopoietic gene expression. [59] [60] [61] [62] [63]

As a part of cohesin complex, functions of Rad21 in the regulation of gene expression include: 1) allele-specific transcription by interacting with the boundary element CCCTC-binding factor (CTCF), [51] [52] [53] [57] [64] [65] 2) tissue-specific transcription by interacting with tissue-specific transcription factors, [53] [66] [67] [68] [69] [70] 3) general progression of transcription by communicating with the basal transcription machinery, [54] [69] [71] [72] and 4) RAD21 co-localization with CTCF-independent pluripotency factors (Oct4, Nanog, Sox4, and KLF2). RAD21 cooperates with CTCF, [73] tissue-specific transcription factors, and basal transcription machinery to regulate transcription dynamically. [74] Also, to effectuate proper transcription activation, cohesin loops chromatin to bring two distant regions together. [65] [70] Cohesin may also act as a transcription insulator to ensure repression. [51] Thus, RAD21 can affect both activation and repression of transcription.  Enhancers that promote transcription and insulators that block transcription are located in conserved regulatory elements (CREs) on chromosomes, and cohesins are thought to physically connect distant CREs with gene promoters in a cell-type specific manner to modulate transcriptional outcome. [75]

In meiosis, REC8 is expressed and replaces RAD21 in the cohesin complex. REC8-containing cohesin generates cohesion between homologous chromosomes and sister chromatids which can persist for years in the case of mammalian oocytes. [76] [77] RAD21L is a further paralog of RAD21 that has a role in meiotic chromosome segregation. [78]  The major role of Rad21L cohesin complex is in homologue pairing and synapsis, not in sister chromatid cohesion, whereas Rec8 most likely functions in sister chromatid cohesion.  Intriguingly, concomitantly with the disappearance of RAD21L, Rad21 appears on the chromosomes in late pachytene and mostly dissociates after diplotene onward. [78] [79] The function of Rad21 cohesin that transiently appears in late prophase I is unclear.

Germline heterozygous or homozygous missense mutations in RAD21 have been associated with human genetic disorders, including developmental diseases such as Cornelia de Lange syndrome [80] [81] [82] [83] [84] [85] [86] [87] [88] [89] [90] and chronic intestinal pseudo-obstruction called Mungan syndrome, [91] [92] respectively, and collectively termed as cohesinopathies.  Somatic mutations and amplification of the RAD21 have also been widely reported in both human solid and hematopoietic tumors. [59] [60] [75] [93] [94] [95] [96] [97] [98] [99] [100] [101] [102] [103] [104] [105] [106] [107] [108] [109] [110] [111] [112] [113]

Notes

Related Research Articles

<span class="mw-page-title-main">Spindle checkpoint</span> Cell cycle checkpoint

The spindle checkpoint, also known as the metaphase-to-anaphase transition, the spindle assembly checkpoint (SAC), the metaphase checkpoint, or the mitotic checkpoint, is a cell cycle checkpoint during metaphase of mitosis or meiosis that prevents the separation of the duplicated chromosomes (anaphase) until each chromosome is properly attached to the spindle. To achieve proper segregation, the two kinetochores on the sister chromatids must be attached to opposite spindle poles. Only this pattern of attachment will ensure that each daughter cell receives one copy of the chromosome. The defining biochemical feature of this checkpoint is the stimulation of the anaphase-promoting complex by M-phase cyclin-CDK complexes, which in turn causes the proteolytic destruction of cyclins and proteins that hold the sister chromatids together.

<span class="mw-page-title-main">Separase</span> Mammalian protein found in Homo sapiens

Separase, also known as separin, is a cysteine protease responsible for triggering anaphase by hydrolysing cohesin, which is the protein responsible for binding sister chromatids during the early stage of anaphase. In humans, separin is encoded by the ESPL1 gene.

<span class="mw-page-title-main">Cohesin</span> Protein complex that regulates the separation of sister chromatids during cell division

Cohesin is a protein complex that mediates sister chromatid cohesion, homologous recombination, and DNA looping. Cohesin is formed of SMC3, SMC1, SCC1 and SCC3. Cohesin holds sister chromatids together after DNA replication until anaphase when removal of cohesin leads to separation of sister chromatids. The complex forms a ring-like structure and it is believed that sister chromatids are held together by entrapment inside the cohesin ring. Cohesin is a member of the SMC family of protein complexes which includes Condensin, MukBEF and SMC-ScpAB.

<span class="mw-page-title-main">CTCF</span> Transcription factor

Transcriptional repressor CTCF also known as 11-zinc finger protein or CCCTC-binding factor is a transcription factor that in humans is encoded by the CTCF gene. CTCF is involved in many cellular processes, including transcriptional regulation, insulator activity, V(D)J recombination and regulation of chromatin architecture.

<span class="mw-page-title-main">SMC1A</span> Protein-coding gene in humans

Structural maintenance of chromosomes protein 1A (SMC1A) is a protein that in humans is encoded by the SMC1A gene. SMC1A is a subunit of the cohesin complex which mediates sister chromatid cohesion, homologous recombination and DNA looping. In somatic cells, cohesin is formed of SMC1A, SMC3, RAD21 and either SA1 or SA2 whereas in meiosis, cohesin is formed of SMC3, SMC1B, REC8 and SA3.

<span class="mw-page-title-main">SMC3</span> Protein-coding gene in humans

Structural maintenance of chromosomes protein 3 (SMC3) is a protein that in humans is encoded by the SMC3 gene. SMC3 is a subunit of the Cohesin complex which mediates sister chromatid cohesion, homologous recombination and DNA looping. Cohesin is formed of SMC3, SMC1, RAD21 and either SA1 or SA2. In humans, SMC3 is present in all cohesin complexes whereas there are multiple paralogs for the other subunits.

<span class="mw-page-title-main">STAG2</span> Protein-coding gene in humans

Cohesin subunit SA-2 (SA2) is a protein that in humans is encoded by the STAG2 gene. SA2 is a subunit of the Cohesin complex which mediates sister chromatid cohesion, homologous recombination and DNA looping. In somatic cells cohesin is formed of SMC3, SMC1, RAD21 and either SA1 or SA2 whereas in meiosis, cohesin is formed of SMC3, SMC1B, REC8 and SA3.

<span class="mw-page-title-main">WAPAL</span> Protein-coding gene in the species Homo sapiens

Wings apart-like protein homolog (WAPL) is a protein that in humans is encoded by the WAPAL gene. WAPL is a key regulator of the Cohesin complex which mediates sister chromatid cohesion, homologous recombination and DNA looping. Cohesin is formed of SMC3, SMC1, RAD21 and either SA1 or SA2. Cohesin has a ring-like arrangement and it is thought that it associates with the chromosome by entrapping it whether as a loop of DNA, a single strand or a pair of sister chromosomes. WAPL forms a complex with PDS5A or PDS5B and releases cohesin from DNA by opening the interface between SMC3 and RAD21.

<span class="mw-page-title-main">PDS5B</span> Protein-coding gene in the species Homo sapiens

Sister chromatid cohesion protein PDS5 homolog B(PDS5B) is a protein that in humans is encoded by the PDS5B gene. It is a regulatory subunit of the Cohesin complex which mediates sister chromatid cohesion, homologous recombination and DNA looping. The core cohesin complex is formed of SMC3, SMC1, RAD21 and either SA1 or SA2. PDS5 associates with WAPL to stimulate the release of cohesin from DNA but during DNA replication PDS5 promotes acetylation of SMC3 by ESCO1 and ESCO2.

<span class="mw-page-title-main">CDCA5</span> Protein-coding gene in the species Homo sapiens

WP:SE

<span class="mw-page-title-main">CTF8</span> Protein-coding gene in the species Homo sapiens

Chromosome transmission fidelity protein 8 homolog is a protein that in humans is encoded by the CHTF8 gene.

<span class="mw-page-title-main">REC8</span> Protein-coding gene in the species Homo sapiens

Meiotic recombination protein REC8 homolog is a protein that in humans is encoded by the REC8 gene.

<span class="mw-page-title-main">STAG1</span> Protein-coding gene in the species Homo sapiens

Cohesin subunit SA-1 (SA1) is a protein that in humans is encoded by the STAG1 gene. SA1 is a subunit of the Cohesin complex which mediates sister chromatid cohesion, homologous recombination and DNA looping. In somatic cells cohesin is formed of SMC3, SMC1, RAD21 and either SA1 or SA2 whereas in meiosis, cohesin is formed of SMC3, SMC1B, REC8 and SA3. There is a nonprofit community formed for those with a STAG1 Gene mutation at www.stag1gene.org.

Sister chromatid cohesion refers to the process by which sister chromatids are paired and held together during certain phases of the cell cycle. Establishment of sister chromatid cohesion is the process by which chromatin-associated cohesin protein becomes competent to physically bind together the sister chromatids. In general, cohesion is established during S phase as DNA is replicated, and is lost when chromosomes segregate during mitosis and meiosis. Some studies have suggested that cohesion aids in aligning the kinetochores during mitosis by forcing the kinetochores to face opposite cell poles.

<span class="mw-page-title-main">SMC1B</span> Protein-coding gene in the species Homo sapiens

Structural maintenance of chromosomes protein 1B (SMC-1B) is a protein that in humans is encoded by the SMC1B gene. SMC proteins engage in chromosome organization and can be broken into 3 groups based on function which are cohesins, condensins, and DNA repair. SMC-1B belongs to a family of proteins required for chromatid cohesion and DNA recombination during meiosis and mitosis. SMC1B protein appears to participate with other cohesins REC8, STAG3 and SMC3 in sister-chromatid cohesion throughout the whole meiotic process in human oocytes.

<span class="mw-page-title-main">STAG3 (gene)</span> Protein-coding gene in the species Homo sapiens

Stromal antigen 3 is a protein that in humans is encoded by the STAG3 gene. STAG3 protein is a component of a cohesin complex that regulates the separation of sister chromatids specifically during meiosis. STAG3 appears to be paramount in sister-chromatid cohesion throughout the meiotic process in human oocytes and spermatocytes.

<span class="mw-page-title-main">Topologically associating domain</span> Self-interacting genomic region

A topologically associating domain (TAD) is a self-interacting genomic region, meaning that DNA sequences within a TAD physically interact with each other more frequently than with sequences outside the TAD. The median size of a TAD in mouse cells is 880 kb, and they have similar sizes in non-mammalian species. Boundaries at both side of these domains are conserved between different mammalian cell types and even across species and are highly enriched with CCCTC-binding factor (CTCF) and cohesin. In addition, some types of genes appear near TAD boundaries more often than would be expected by chance.

<span class="mw-page-title-main">Frank Uhlmann</span>

Frank Uhlmann FRS is a group leader at the Francis Crick Institute in London.

<span class="mw-page-title-main">Nuclear organization</span> Spatial distribution of chromatin within a cell nucleus

Nuclear organization refers to the spatial distribution of chromatin within a cell nucleus. There are many different levels and scales of nuclear organisation. Chromatin is a higher order structure of DNA.

<span class="mw-page-title-main">Insulated neighborhood</span>

In mammalian biology, insulated neighborhoods are chromosomal loop structures formed by the physical interaction of two DNA loci bound by the transcription factor CTCF and co-occupied by cohesin. Insulated neighborhoods are thought to be structural and functional units of gene control because their integrity is important for normal gene regulation. Current evidence suggests that these structures form the mechanistic underpinnings of higher-order chromosome structures, including topologically associating domains (TADs). Insulated neighborhoods are functionally important in understanding gene regulation in normal cells and dysregulated gene expression in disease.

References

  1. 1 2 3 GRCh38: Ensembl release 89: ENSG00000164754 - Ensembl, May 2017
  2. 1 2 3 GRCm38: Ensembl release 89: ENSMUSG00000022314 - Ensembl, May 2017
  3. "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. McKay MJ, Troelstra C, van der Spek P, et al. (Jan 1997). "Sequence conservation of the rad21 Schizosaccharomyces pombe DNA double-strand break repair gene in human and mouse". Genomics. 36 (2): 305–15. doi:10.1006/geno.1996.0466. hdl: 1765/3107 . PMID   8812457.
  6. "Entrez Gene: RAD21 RAD21 homolog (S. pombe)".
  7. Birkenbihl RP, Subramani S (December 1992). "Cloning and characterization of rad21 an essential gene of Schizosaccharomyces pombe involved in DNA double-strand-break repair". Nucleic Acids Research. 20 (24): 6605–11. doi:10.1093/nar/20.24.6605. PMC   334577 . PMID   1480481.
  8. 1 2 McKay MJ, Troelstra C, van der Spek P, et al. (September 1996). "Sequence conservation of the rad21 Schizosaccharomyces pombe DNA double-strand break repair gene in human and mouse". Genomics. 36 (2): 305–15. doi:10.1006/geno.1996.0466. hdl: 1765/3107 . PMID   8812457.
  9. Nomura N, Nagase T, Miyajima N, et al. (1994-01-01). "Prediction of the coding sequences of unidentified human genes. II. The coding sequences of 40 new genes (KIAA0041-KIAA0080) deduced by analysis of cDNA clones from human cell line KG-1". DNA Research. 1 (5): 223–9. doi: 10.1093/dnares/1.5.223 . PMID   7584044.
  10. Guacci V, Koshland D, Strunnikov A (October 1997). "A direct link between sister chromatid cohesion and chromosome condensation revealed through the analysis of MCD1 in S. cerevisiae". Cell. 91 (1): 47–57. doi:10.1016/S0092-8674(01)80008-8. PMC   2670185 . PMID   9335334.
  11. Michaelis C, Ciosk R, Nasmyth K (October 1997). "Cohesins: chromosomal proteins that prevent premature separation of sister chromatids". Cell. 91 (1): 35–45. doi: 10.1016/S0092-8674(01)80007-6 . PMID   9335333. S2CID   18572651.
  12. 1 2 3 Uhlmann F, Lottspeich F, Nasmyth K (July 1999). "Sister-chromatid separation at anaphase onset is promoted by cleavage of the cohesin subunit Scc1". Nature. 400 (6739): 37–42. Bibcode:1999Natur.400...37U. doi:10.1038/21831. PMID   10403247. S2CID   4354549.
  13. Nasmyth K, Haering CH (June 2005). "The structure and function of SMC and kleisin complexes". Annual Review of Biochemistry. 74 (1): 595–648. doi:10.1146/annurev.biochem.74.082803.133219. PMID   15952899.
  14. 1 2 Hauf S, Waizenegger IC, Peters JM (August 2001). "Cohesin cleavage by separase required for anaphase and cytokinesis in human cells". Science. 293 (5533): 1320–3. Bibcode:2001Sci...293.1320H. doi:10.1126/science.1061376. PMID   11509732. S2CID   46036132.
  15. Uhlmann F, Wernic D, Poupart MA, et al. (October 2000). "Cleavage of cohesin by the CD clan protease separin triggers anaphase in yeast". Cell. 103 (3): 375–86. doi: 10.1016/S0092-8674(00)00130-6 . PMID   11081625. S2CID   2667617.
  16. 1 2 Panigrahi AK, Zhang N, Mao Q, et al. (November 2011). "Calpain-1 cleaves Rad21 to promote sister chromatid separation". Molecular and Cellular Biology. 31 (21): 4335–47. doi:10.1128/MCB.06075-11. PMC   3209327 . PMID   21876002.
  17. 1 2 3 Chen F, Kamradt M, Mulcahy M, et al. (May 2002). "Caspase proteolysis of the cohesin component RAD21 promotes apoptosis". The Journal of Biological Chemistry. 277 (19): 16775–81. doi: 10.1074/jbc.M201322200 . PMID   11875078.
  18. 1 2 3 Pati D, Zhang N, Plon SE (December 2002). "Linking sister chromatid cohesion and apoptosis: role of Rad21". Molecular and Cellular Biology. 22 (23): 8267–77. doi:10.1128/MCB.22.23.8267-8277.2002. PMC   134054 . PMID   12417729.
  19. Kosugi S, Hasebe M, Tomita M, et al. (June 2009). "Systematic identification of cell cycle-dependent yeast nucleocytoplasmic shuttling proteins by prediction of composite motifs". Proceedings of the National Academy of Sciences of the United States of America. 106 (25): 10171–6. Bibcode:2009PNAS..10610171K. doi: 10.1073/pnas.0900604106 . PMC   2695404 . PMID   19520826.
  20. 1 2 Gligoris TG, Scheinost JC, Bürmann F, et al. (November 2014). "Closing the cohesin ring: structure and function of its Smc3-kleisin interface". Science. 346 (6212): 963–7. Bibcode:2014Sci...346..963G. doi:10.1126/science.1256917. PMC   4300515 . PMID   25414305.
  21. Haering CH, Löwe J, Hochwagen A, et al. (April 2002). "Molecular architecture of SMC proteins and the yeast cohesin complex". Molecular Cell. 9 (4): 773–88. doi: 10.1016/S1097-2765(02)00515-4 . PMID   11983169.
  22. 1 2 Zhang N, Kuznetsov SG, Sharan SK, et al. (December 2008). "A handcuff model for the cohesin complex". The Journal of Cell Biology. 183 (6): 1019–31. doi:10.1083/jcb.200801157. PMC   2600748 . PMID   19075111.
  23. Zhang N, Pati D (February 2009). "Handcuff for sisters: a new model for sister chromatid cohesion". Cell Cycle. 8 (3): 399–402. doi:10.4161/cc.8.3.7586. PMC   2689371 . PMID   19177018.
  24. Zhang N, Pati D (June 2012). "Sororin is a master regulator of sister chromatid cohesion and separation". Cell Cycle. 11 (11): 2073–83. doi:10.4161/cc.20241. PMC   3368859 . PMID   22580470.
  25. Nishiyama T, Ladurner R, Schmitz J, et al. (November 2010). "Sororin mediates sister chromatid cohesion by antagonizing Wapl". Cell. 143 (5): 737–49. doi: 10.1016/j.cell.2010.10.031 . PMID   21111234. S2CID   518782.
  26. 1 2 Zhang N, Pati D (2014). "Road to cancer via cohesin deregulation.". Oncology: Theory & Practice. iConcept Press Hong Kong. pp. 213–240.
  27. Hara K, Zheng G, Qu Q, et al. (October 2014). "Structure of cohesin subcomplex pinpoints direct shugoshin-Wapl antagonism in centromeric cohesion". Nature Structural & Molecular Biology. 21 (10): 864–70. doi:10.1038/nsmb.2880. PMC   4190070 . PMID   25173175.
  28. Petela NJ, Gligoris TG, Metson J, et al. (June 2018). "Scc2 Is a Potent Activator of Cohesin's ATPase that Promotes Loading by Binding Scc1 without Pds5". Molecular Cell. 70 (6): 1134–1148.e7. doi:10.1016/j.molcel.2018.05.022. PMC   6028919 . PMID   29932904.
  29. Kikuchi S, Borek DM, Otwinowski Z, et al. (November 2016). "Crystal structure of the cohesin loader Scc2 and insight into cohesinopathy". Proceedings of the National Academy of Sciences of the United States of America. 113 (44): 12444–12449. Bibcode:2016PNAS..11312444K. doi: 10.1073/pnas.1611333113 . PMC   5098657 . PMID   27791135.
  30. Muir KW, Kschonsak M, Li Y, et al. (March 2016). "Structure of the Pds5-Scc1 Complex and Implications for Cohesin Function". Cell Reports. 14 (9): 2116–2126. doi: 10.1016/j.celrep.2016.01.078 . PMID   26923589.
  31. Haering CH, Schoffnegger D, Nishino T, et al. (September 2004). "Structure and stability of cohesin's Smc1-kleisin interaction". Molecular Cell. 15 (6): 951–64. doi: 10.1016/j.molcel.2004.08.030 . PMID   15383284.
  32. Beckouët F, Srinivasan M, Roig MB, et al. (February 2016). "Releasing Activity Disengages Cohesin's Smc3/Scc1 Interface in a Process Blocked by Acetylation". Molecular Cell. 61 (4): 563–574. doi:10.1016/j.molcel.2016.01.026. PMC   4769318 . PMID   26895425.
  33. Muir KW, Li Y, Weis F, et al. (March 2020). "The structure of the cohesin ATPase elucidates the mechanism of SMC-kleisin ring opening". Nature Structural & Molecular Biology. 27 (3): 233–239. doi:10.1038/s41594-020-0379-7. PMC   7100847 . PMID   32066964.
  34. "RAD21 cohesin complex component [Homo sapiens]". NCBI gene. National Center for Biotechnology Information (NCBI), U.S. National Library of Medicine.
  35. Panigrahi AK, Zhang N, Otta SK, et al. (March 2012). "A cohesin-RAD21 interactome". The Biochemical Journal. 442 (3): 661–70. doi:10.1042/BJ20111745. PMID   22145905. S2CID   46097282.
  36. Gregson HC, Schmiesing JA, Kim JS, et al. (December 2001). "A potential role for human cohesin in mitotic spindle aster assembly". The Journal of Biological Chemistry. 276 (50): 47575–82. doi: 10.1074/jbc.M103364200 . PMID   11590136.
  37. Guillou E, Ibarra A, Coulon V, et al. (December 2010). "Cohesin organizes chromatin loops at DNA replication factories". Genes & Development. 24 (24): 2812–22. doi:10.1101/gad.608210. PMC   3003199 . PMID   21159821.
  38. Takahashi TS, Yiu P, Chou MF, et al. (October 2004). "Recruitment of Xenopus Scc2 and cohesin to chromatin requires the pre-replication complex". Nature Cell Biology. 6 (10): 991–6. doi:10.1038/ncb1177. PMID   15448702. S2CID   20488928.
  39. Ryu MJ, Kim BJ, Lee JW, et al. (March 2006). "Direct interaction between cohesin complex and DNA replication machinery". Biochemical and Biophysical Research Communications. 341 (3): 770–5. doi:10.1016/j.bbrc.2006.01.029. PMID   16438930.
  40. Terret ME, Sherwood R, Rahman S, et al. (November 2009). "Cohesin acetylation speeds the replication fork". Nature. 462 (7270): 231–4. Bibcode:2009Natur.462..231T. doi:10.1038/nature08550. PMC   2777716 . PMID   19907496.
  41. MacAlpine HK, Gordân R, Powell SK, et al. (February 2010). "Drosophila ORC localizes to open chromatin and marks sites of cohesin complex loading". Genome Research. 20 (2): 201–11. doi:10.1101/gr.097873.109. PMC   2813476 . PMID   19996087.
  42. Unal E, Heidinger-Pauli JM, Koshland D (July 2007). "DNA double-strand breaks trigger genome-wide sister-chromatid cohesion through Eco1 (Ctf7)". Science. 317 (5835): 245–8. Bibcode:2007Sci...317..245U. doi:10.1126/science.1140637. PMID   17626885. S2CID   551399.
  43. Heidinger-Pauli JM, Unal E, Koshland D (May 2009). "Distinct targets of the Eco1 acetyltransferase modulate cohesion in S phase and in response to DNA damage". Molecular Cell. 34 (3): 311–21. doi:10.1016/j.molcel.2009.04.008. PMC   2737744 . PMID   19450529.
  44. Ström L, Lindroos HB, Shirahige K, et al. (December 2004). "Postreplicative recruitment of cohesin to double-strand breaks is required for DNA repair". Molecular Cell. 16 (6): 1003–15. doi: 10.1016/j.molcel.2004.11.026 . PMID   15610742.
  45. Kim BJ, Li Y, Zhang J, et al. (July 2010). "Genome-wide reinforcement of cohesin binding at pre-existing cohesin sites in response to ionizing radiation in human cells". The Journal of Biological Chemistry. 285 (30): 22784–92. doi: 10.1074/jbc.M110.134577 . PMC   2906269 . PMID   20501661.
  46. Watrin E, Peters JM (September 2009). "The cohesin complex is required for the DNA damage-induced G2/M checkpoint in mammalian cells". The EMBO Journal. 28 (17): 2625–35. doi:10.1038/emboj.2009.202. PMC   2738698 . PMID   19629043.
  47. Cortés-Ledesma F, Aguilera A (September 2006). "Double-strand breaks arising by replication through a nick are repaired by cohesin-dependent sister-chromatid exchange". EMBO Reports. 7 (9): 919–26. doi:10.1038/sj.embor.7400774. PMC   1559660 . PMID   16888651.
  48. Watrin E, Peters JM (August 2006). "Cohesin and DNA damage repair". Experimental Cell Research. 312 (14): 2687–93. doi:10.1016/j.yexcr.2006.06.024. PMID   16876157.
  49. Ball AR, Yokomori K (January 2008). "Damage-induced reactivation of cohesin in postreplicative DNA repair". BioEssays. 30 (1): 5–9. doi:10.1002/bies.20691. PMC   4127326 . PMID   18081005.
  50. Sjögren C, Ström L (May 2010). "S-phase and DNA damage activated establishment of sister chromatid cohesion--importance for DNA repair". Experimental Cell Research. 316 (9): 1445–53. doi:10.1016/j.yexcr.2009.12.018. PMID   20043905.
  51. 1 2 3 Wendt KS, Yoshida K, Itoh T, et al. (February 2008). "Cohesin mediates transcriptional insulation by CCCTC-binding factor". Nature. 451 (7180): 796–801. Bibcode:2008Natur.451..796W. doi:10.1038/nature06634. PMID   18235444. S2CID   205212289.
  52. 1 2 Skibbens RV, Marzillier J, Eastman L (April 2010). "Cohesins coordinate gene transcriptions of related function within Saccharomyces cerevisiae". Cell Cycle. 9 (8): 1601–6. doi:10.4161/cc.9.8.11307. PMC   3096706 . PMID   20404480.
  53. 1 2 3 Schmidt D, Schwalie PC, Ross-Innes CS, et al. (May 2010). "A CTCF-independent role for cohesin in tissue-specific transcription". Genome Research. 20 (5): 578–88. doi:10.1101/gr.100479.109. PMC   2860160 . PMID   20219941.
  54. 1 2 Kagey MH, Newman JJ, Bilodeau S, et al. (September 2010). "Mediator and cohesin connect gene expression and chromatin architecture". Nature. 467 (7314): 430–5. Bibcode:2010Natur.467..430K. doi:10.1038/nature09380. PMC   2953795 . PMID   20720539.
  55. Pauli A, van Bemmel JG, Oliveira RA, et al. (October 2010). "A direct role for cohesin in gene regulation and ecdysone response in Drosophila salivary glands". Current Biology. 20 (20): 1787–98. Bibcode:2010CBio...20.1787P. doi:10.1016/j.cub.2010.09.006. PMC   4763543 . PMID   20933422.
  56. Dorsett D (October 2010). "Gene regulation: the cohesin ring connects developmental highways". Current Biology. 20 (20): R886-8. Bibcode:2010CBio...20.R886D. doi: 10.1016/j.cub.2010.09.036 . PMID   20971431. S2CID   2543711.
  57. 1 2 Parelho V, Hadjur S, Spivakov M, et al. (February 2008). "Cohesins functionally associate with CTCF on mammalian chromosome arms". Cell. 132 (3): 422–33. doi: 10.1016/j.cell.2008.01.011 . PMID   18237772. S2CID   14363394.
  58. Liu J, Zhang Z, Bando M, et al. (May 2009). Hastie N (ed.). "Transcriptional dysregulation in NIPBL and cohesin mutant human cells". PLOS Biology. 7 (5): e1000119. doi: 10.1371/journal.pbio.1000119 . PMC   2680332 . PMID   19468298.
  59. 1 2 Mazumdar C, Shen Y, Xavy S, et al. (December 2015). "Leukemia-Associated Cohesin Mutants Dominantly Enforce Stem Cell Programs and Impair Human Hematopoietic Progenitor Differentiation". Cell Stem Cell. 17 (6): 675–688. doi:10.1016/j.stem.2015.09.017. PMC   4671831 . PMID   26607380.
  60. 1 2 Mullenders J, Aranda-Orgilles B, Lhoumaud P, et al. (October 2015). "Cohesin loss alters adult hematopoietic stem cell homeostasis, leading to myeloproliferative neoplasms". The Journal of Experimental Medicine. 212 (11): 1833–50. doi:10.1084/jem.20151323. PMC   4612095 . PMID   26438359.
  61. Viny AD, Ott CJ, Spitzer B, et al. (October 2015). "Dose-dependent role of the cohesin complex in normal and malignant hematopoiesis". The Journal of Experimental Medicine. 212 (11): 1819–32. doi:10.1084/jem.20151317. PMC   4612085 . PMID   26438361.
  62. Fisher JB, McNulty M, Burke MJ, et al. (April 2017). "Cohesin Mutations in Myeloid Malignancies". Trends in Cancer. 3 (4): 282–293. doi:10.1016/j.trecan.2017.02.006. PMC   5472227 . PMID   28626802.
  63. Rao S (December 2019). "Closing the loop on cohesin in hematopoiesis". Blood. 134 (24): 2123–2125. doi:10.1182/blood.2019003279. PMC   6908834 . PMID   31830276.
  64. Degner SC, Verma-Gaur J, Wong TP, et al. (June 2011). "CCCTC-binding factor (CTCF) and cohesin influence the genomic architecture of the Igh locus and antisense transcription in pro-B cells". Proceedings of the National Academy of Sciences of the United States of America. 108 (23): 9566–71. Bibcode:2011PNAS..108.9566D. doi: 10.1073/pnas.1019391108 . PMC   3111298 . PMID   21606361.
  65. 1 2 Guo Y, Monahan K, Wu H, et al. (December 2012). "CTCF/cohesin-mediated DNA looping is required for protocadherin α promoter choice". Proceedings of the National Academy of Sciences of the United States of America. 109 (51): 21081–6. Bibcode:2012PNAS..10921081G. doi: 10.1073/pnas.1219280110 . PMC   3529044 . PMID   23204437.
  66. Hadjur S, Williams LM, Ryan NK, et al. (July 2009). "Cohesins form chromosomal cis-interactions at the developmentally regulated IFNG locus". Nature. 460 (7253): 410–3. Bibcode:2009Natur.460..410H. doi:10.1038/nature08079. PMC   2869028 . PMID   19458616.
  67. Faure AJ, Schmidt D, Watt S, et al. (November 2012). "Cohesin regulates tissue-specific expression by stabilizing highly occupied cis-regulatory modules". Genome Research. 22 (11): 2163–75. doi:10.1101/gr.136507.111. PMC   3483546 . PMID   22780989.
  68. Seitan VC, Hao B, Tachibana-Konwalski K, et al. (August 2011). "A role for cohesin in T-cell-receptor rearrangement and thymocyte differentiation". Nature. 476 (7361): 467–71. Bibcode:2011Natur.476..467S. doi:10.1038/nature10312. PMC   3179485 . PMID   21832993.
  69. 1 2 Yan J, Enge M, Whitington T, et al. (August 2013). "Transcription factor binding in human cells occurs in dense clusters formed around cohesin anchor sites". Cell. 154 (4): 801–13. doi: 10.1016/j.cell.2013.07.034 . PMID   23953112.
  70. 1 2 Zhang H, Jiao W, Sun L, et al. (July 2013). "Intrachromosomal looping is required for activation of endogenous pluripotency genes during reprogramming". Cell Stem Cell. 13 (1): 30–5. doi: 10.1016/j.stem.2013.05.012 . PMID   23747202.
  71. Fay A, Misulovin Z, Li J, et al. (October 2011). "Cohesin selectively binds and regulates genes with paused RNA polymerase". Current Biology. 21 (19): 1624–34. Bibcode:2011CBio...21.1624F. doi:10.1016/j.cub.2011.08.036. PMC   3193539 . PMID   21962715.
  72. Schaaf CA, Kwak H, Koenig A, et al. (March 2013). Ren B (ed.). "Genome-wide control of RNA polymerase II activity by cohesin". PLOS Genetics. 9 (3): e1003382. doi: 10.1371/journal.pgen.1003382 . PMC   3605059 . PMID   23555293.
  73. Rubio ED, Reiss DJ, Welcsh PL, et al. (June 2008). "CTCF physically links cohesin to chromatin". Proceedings of the National Academy of Sciences of the United States of America. 105 (24): 8309–14. Bibcode:2008PNAS..105.8309R. doi: 10.1073/pnas.0801273105 . PMC   2448833 . PMID   18550811.
  74. Dorsett D, Merkenschlager M (June 2013). "Cohesin at active genes: a unifying theme for cohesin and gene expression from model organisms to humans". Current Opinion in Cell Biology. 25 (3): 327–33. doi:10.1016/j.ceb.2013.02.003. PMC   3691354 . PMID   23465542.
  75. 1 2 Leeke B, Marsman J, O'Sullivan JM, et al. (2014). "Cohesin mutations in myeloid malignancies: underlying mechanisms". Experimental Hematology & Oncology. 3 (1): 13. doi: 10.1186/2162-3619-3-13 . PMC   4046106 . PMID   24904756.
  76. Tachibana-Konwalski K, Godwin J, van der Weyden L, et al. (November 2010). "Rec8-containing cohesin maintains bivalents without turnover during the growing phase of mouse oocytes". Genes & Development. 24 (22): 2505–16. doi:10.1101/gad.605910. PMC   2975927 . PMID   20971813.
  77. Buonomo SB, Clyne RK, Fuchs J, et al. (October 2000). "Disjunction of homologous chromosomes in meiosis I depends on proteolytic cleavage of the meiotic cohesin Rec8 by separin". Cell. 103 (3): 387–98. doi: 10.1016/S0092-8674(00)00131-8 . PMID   11081626. S2CID   17385055.
  78. 1 2 Lee J, Hirano T (January 2011). "RAD21L, a novel cohesin subunit implicated in linking homologous chromosomes in mammalian meiosis". The Journal of Cell Biology. 192 (2): 263–76. doi:10.1083/jcb.201008005. PMC   3172173 . PMID   21242291.
  79. Ishiguro K, Kim J, Fujiyama-Nakamura S, et al. (March 2011). "A new meiosis-specific cohesin complex implicated in the cohesin code for homologous pairing". EMBO Reports. 12 (3): 267–75. doi:10.1038/embor.2011.2. PMC   3059921 . PMID   21274006.
  80. Krab LC, Marcos-Alcalde I, Assaf M, et al. (May 2020). "Delineation of phenotypes and genotypes related to cohesin structural protein RAD21". Human Genetics. 139 (5): 575–592. doi:10.1007/s00439-020-02138-2. PMC   7170815 . PMID   32193685.
  81. Deardorff MA, Wilde JJ, Albrecht M, et al. (June 2012). "RAD21 mutations cause a human cohesinopathy". American Journal of Human Genetics. 90 (6): 1014–27. doi:10.1016/j.ajhg.2012.04.019. PMC   3370273 . PMID   22633399.
  82. Ansari M, Poke G, Ferry Q, et al. (October 2014). "Genetic heterogeneity in Cornelia de Lange syndrome (CdLS) and CdLS-like phenotypes with observed and predicted levels of mosaicism". Journal of Medical Genetics. 51 (10): 659–68. doi:10.1136/jmedgenet-2014-102573. PMC   4173748 . PMID   25125236.
  83. Minor A, Shinawi M, Hogue JS, et al. (March 2014). "Two novel RAD21 mutations in patients with mild Cornelia de Lange syndrome-like presentation and report of the first familial case". Gene. 537 (2): 279–84. doi:10.1016/j.gene.2013.12.045. PMID   24378232.
  84. Boyle MI, Jespersgaard C, Nazaryan L, et al. (April 2017). "A novel RAD21 variant associated with intrafamilial phenotypic variation in Cornelia de Lange syndrome - review of the literature". Clinical Genetics. 91 (4): 647–649. doi:10.1111/cge.12863. PMID   27882533. S2CID   3732288.
  85. Martínez F, Caro-Llopis A, Roselló M, et al. (February 2017). "High diagnostic yield of syndromic intellectual disability by targeted next-generation sequencing". Journal of Medical Genetics. 54 (2): 87–92. doi:10.1136/jmedgenet-2016-103964. PMID   27620904. S2CID   46740644.
  86. Dorval S, Masciadri M, Mathot M, et al. (January 2020). "A novel RAD21 mutation in a boy with mild Cornelia de Lange presentation: Further delineation of the phenotype". European Journal of Medical Genetics. 63 (1): 103620. doi:10.1016/j.ejmg.2019.01.010. PMID   30716475. S2CID   73443712.
  87. Gudmundsson S, Annerén G, Marcos-Alcalde Í, et al. (June 2019). "A novel RAD21 p.(Gln592del) variant expands the clinical description of Cornelia de Lange syndrome type 4 - Review of the literature". European Journal of Medical Genetics. 62 (6): 103526. doi:10.1016/j.ejmg.2018.08.007. hdl: 10641/1986 . PMID   30125677. S2CID   52046223.
  88. Pereza N, Severinski S, Ostojić S, et al. (June 2015). "Cornelia de Lange syndrome caused by heterozygous deletions of chromosome 8q24: comments on the article by Pereza et al. [2012]". American Journal of Medical Genetics. Part A. 167 (6): 1426–7. doi:10.1002/ajmg.a.36974. PMID   25899858. S2CID   205320077.
  89. Wuyts W, Roland D, Lüdecke HJ, et al. (December 2002). "Multiple exostoses, mental retardation, hypertrichosis, and brain abnormalities in a boy with a de novo 8q24 submicroscopic interstitial deletion". American Journal of Medical Genetics. 113 (4): 326–32. doi:10.1002/ajmg.10845. PMID   12457403.
  90. McBrien J, Crolla JA, Huang S, et al. (June 2008). "Further case of microdeletion of 8q24 with phenotype overlapping Langer-Giedion without TRPS1 deletion". American Journal of Medical Genetics. Part A. 146A (12): 1587–92. doi:10.1002/ajmg.a.32347. PMID   18478595. S2CID   19384557.
  91. Bonora E, Bianco F, Cordeddu L, et al. (April 2015). "Mutations in RAD21 disrupt regulation of APOB in patients with chronic intestinal pseudo-obstruction". Gastroenterology. 148 (4): 771–782.e11. doi:10.1053/j.gastro.2014.12.034. hdl:11693/23636. PMC   4375026 . PMID   25575569.
  92. Mungan Z, Akyüz F, Bugra Z, et al. (November 2003). "Familial visceral myopathy with pseudo-obstruction, megaduodenum, Barrett's esophagus, and cardiac abnormalities". The American Journal of Gastroenterology. 98 (11): 2556–60. doi:10.1111/j.1572-0241.2003.08707.x. PMID   14638363. S2CID   21022551.
  93. Mintzas K, Heuser M (June 2019). "Emerging strategies to target the dysfunctional cohesin complex in cancer". Expert Opinion on Therapeutic Targets. 23 (6): 525–537. doi:10.1080/14728222.2019.1609943. PMID   31020869. S2CID   131776323.
  94. van 't Veer LJ, Dai H, van de Vijver MJ, et al. (January 2002). "Gene expression profiling predicts clinical outcome of breast cancer". Nature. 415 (6871): 530–6. doi:10.1038/415530a. hdl: 1874/15552 . PMID   11823860. S2CID   4369266.
  95. Xu H, Yan M, Patra J, et al. (January 2011). "Enhanced RAD21 cohesin expression confers poor prognosis and resistance to chemotherapy in high grade luminal, basal and HER2 breast cancers". Breast Cancer Research. 13 (1): R9. doi: 10.1186/bcr2814 . PMC   3109576 . PMID   21255398.
  96. Yamamoto G, Irie T, Aida T, et al. (April 2006). "Correlation of invasion and metastasis of cancer cells, and expression of the RAD21 gene in oral squamous cell carcinoma". Virchows Archiv. 448 (4): 435–41. doi:10.1007/s00428-005-0132-y. PMID   16416296. S2CID   22993345.
  97. Deb S, Xu H, Tuynman J, et al. (March 2014). "RAD21 cohesin overexpression is a prognostic and predictive marker exacerbating poor prognosis in KRAS mutant colorectal carcinomas". British Journal of Cancer. 110 (6): 1606–13. doi:10.1038/bjc.2014.31. PMC   3960611 . PMID   24548858.
  98. Porkka KP, Tammela TL, Vessella RL, et al. (January 2004). "RAD21 and KIAA0196 at 8q24 are amplified and overexpressed in prostate cancer". Genes, Chromosomes & Cancer. 39 (1): 1–10. doi: 10.1002/gcc.10289 . PMID   14603436. S2CID   46570803.
  99. Yun J, Song SH, Kang JY, et al. (January 2016). "Reduced cohesin destabilizes high-level gene amplification by disrupting pre-replication complex bindings in human cancers with chromosomal instability". Nucleic Acids Research. 44 (2): 558–72. doi:10.1093/nar/gkv933. PMC   4737181 . PMID   26420833.
  100. Fisher JB, Peterson J, Reimer M, et al. (March 2017). "The cohesin subunit Rad21 is a negative regulator of hematopoietic self-renewal through epigenetic repression of Hoxa7 and Hoxa9". Leukemia. 31 (3): 712–719. doi:10.1038/leu.2016.240. PMC   5332284 . PMID   27554164.
  101. Solomon DA, Kim JS, Waldman T (June 2014). "Cohesin gene mutations in tumorigenesis: from discovery to clinical significance". BMB Reports. 47 (6): 299–310. doi:10.5483/BMBRep.2014.47.6.092. PMC   4163871 . PMID   24856830.
  102. Thota S, Viny AD, Makishima H, et al. (September 2014). "Genetic alterations of the cohesin complex genes in myeloid malignancies". Blood. 124 (11): 1790–8. doi:10.1182/blood-2014-04-567057. PMC   4162108 . PMID   25006131.
  103. Hill VK, Kim JS, Waldman T (August 2016). "Cohesin mutations in human cancer". Biochimica et Biophysica Acta (BBA) - Reviews on Cancer. 1866 (1): 1–11. doi:10.1016/j.bbcan.2016.05.002. PMC   4980180 . PMID   27207471.
  104. Corces-Zimmerman MR, Hong WJ, Weissman IL, et al. (February 2014). "Preleukemic mutations in human acute myeloid leukemia affect epigenetic regulators and persist in remission". Proceedings of the National Academy of Sciences of the United States of America. 111 (7): 2548–53. Bibcode:2014PNAS..111.2548C. doi: 10.1073/pnas.1324297111 . PMC   3932921 . PMID   24550281.
  105. Ley TJ, Miller C, Ding L, et al. (May 2013). "Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia". The New England Journal of Medicine. 368 (22): 2059–74. doi:10.1056/NEJMoa1301689. PMC   3767041 . PMID   23634996.
  106. Kon A, Shih LY, Minamino M, et al. (October 2013). "Recurrent mutations in multiple components of the cohesin complex in myeloid neoplasms". Nature Genetics. 45 (10): 1232–7. doi:10.1038/ng.2731. PMID   23955599. S2CID   12395243.
  107. Thol F, Bollin R, Gehlhaar M, et al. (February 2014). "Mutations in the cohesin complex in acute myeloid leukemia: clinical and prognostic implications". Blood. 123 (6): 914–20. doi: 10.1182/blood-2013-07-518746 . PMID   24335498. S2CID   206923475.
  108. Lindsley RC, Mar BG, Mazzola E, et al. (February 2015). "Acute myeloid leukemia ontogeny is defined by distinct somatic mutations". Blood. 125 (9): 1367–76. doi:10.1182/blood-2014-11-610543. PMC   4342352 . PMID   25550361.
  109. Tsai CH, Hou HA, Tang JL, et al. (December 2017). "Prognostic impacts and dynamic changes of cohesin complex gene mutations in de novo acute myeloid leukemia". Blood Cancer Journal. 7 (12): 663. doi:10.1038/s41408-017-0022-y. PMC   5802563 . PMID   29288251.
  110. Eisfeld AK, Kohlschmidt J, Mrózek K, et al. (June 2018). "Mutation patterns identify adult patients with de novo acute myeloid leukemia aged 60 years or older who respond favorably to standard chemotherapy: an analysis of Alliance studies". Leukemia. 32 (6): 1338–1348. doi:10.1038/s41375-018-0068-2. PMC   5992022 . PMID   29563537.
  111. Weinberg OK, Gibson CJ, Blonquist TM, et al. (April 2018). "de novo acute myeloid leukemia without 2016 WHO Classification-defined cytogenetic abnormalities". Haematologica. 103 (4): 626–633. doi:10.3324/haematol.2017.181842. PMC   5865424 . PMID   29326119.
  112. Duployez N, Marceau-Renaut A, Boissel N, et al. (May 2016). "Comprehensive mutational profiling of core binding factor acute myeloid leukemia". Blood. 127 (20): 2451–9. doi:10.1182/blood-2015-12-688705. PMC   5457131 . PMID   26980726.
  113. Yoshida K, Toki T, Okuno Y, et al. (November 2013). "The landscape of somatic mutations in Down syndrome-related myeloid disorders". Nature Genetics. 45 (11): 1293–9. doi:10.1038/ng.2759. PMID   24056718. S2CID   32383374.