fbpx
Wikipedia

CDKN1B

Cyclin-dependent kinase inhibitor 1B (p27Kip1) is an enzyme inhibitor that in humans is encoded by the CDKN1B gene.[5] It encodes a protein which belongs to the Cip/Kip family of cyclin dependent kinase (Cdk) inhibitor proteins. The encoded protein binds to and prevents the activation of cyclin E-CDK2 or cyclin D-CDK4 complexes, and thus controls the cell cycle progression at G1. It is often referred to as a cell cycle inhibitor protein because its major function is to stop or slow down the cell division cycle.

CDKN1B
Available structures
PDBOrtholog search: PDBe RCSB
Identifiers
AliasesCDKN1B, CDKN4, KIP1, MEN1B, MEN4, P27KIP1, cyclin-dependent kinase inhibitor 1B, cyclin dependent kinase inhibitor 1B
External IDsOMIM: 600778 MGI: 104565 HomoloGene: 2999 GeneCards: CDKN1B
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_004064

NM_009875

RefSeq (protein)

NP_004055

NP_034005

Location (UCSC)Chr 12: 12.69 – 12.72 MbChr 6: 134.9 – 134.9 Mb
PubMed search[3][4]
Wikidata
View/Edit HumanView/Edit Mouse

Function edit

The p27Kip1 gene has a DNA sequence similar to other members of the "Cip/Kip" family which include the p21Cip1/Waf1 and p57Kip2 genes. In addition to this structural similarity the "Cip/Kip" proteins share the functional characteristic of being able to bind several different classes of Cyclin and Cdk molecules. For example, p27Kip1 binds to cyclin D either alone, or when complexed to its catalytic subunit CDK4. In doing so p27Kip1 inhibits the catalytic activity of Cdk4, which means that it prevents Cdk4 from adding phosphate residues to its principal substrate, the retinoblastoma (pRb) protein. Increased levels of the p27Kip1 protein typically cause cells to arrest in the G1 phase of the cell cycle. Likewise, p27Kip1 is able to bind other Cdk proteins when complexed to cyclin subunits such as Cyclin E/Cdk2 and Cyclin A/Cdk2.[6]

Regulation edit

In general, extracellular growth factors which promote cell division reduce transcription and translation of p27Kip1. Also, increased synthesis of CDk4,6/cyclin D causes binding of p27 to this complex, sequestering it from binding to the CDk2/cyclin E complex. Furthermore, an active CDK2/cyclin E complex will phosphorylate p27 and tag p27 for ubiquitination.[7] A mutation of this gene may lead to loss of control over the cell cycle leading to uncontrolled cellular proliferation.[8][9][10] Loss of p27 expression has been observed in metastatic canine mammary carcinomas.[11][12][13] Decreased TGF-beta signalling has been suggested to cause loss of p27 expression in this tumor type.[14]

A structured cis-regulatory element has been found in the 5' UTR of the P27 mRNA where it is thought to regulate translation relative to cell cycle progression.[15]

P27 regulation is accomplished by two different mechanisms. In the first its concentration is changed by the individual rates of transcription, translation, and proteolysis. P27 can also be regulated by changing its subcellular location [16] Both mechanisms act to reduce levels of p27, allowing for the activation of Cdk1 and Cdk2, and for the cell to begin progressing through the cell cycle.

Transcription edit

Transcription of the CDKN1B gene is activated by Forkhead box class O family (FoxO) proteins which also acts downstream to promote p27 nuclear localization and decrease levels of COP9 subunit 5(COPS5) which helps in the degradation of p27.[17] Transcription for p27 is activated by FoxO in response to cytokines, promyelocytic leukaemia proteins, and nuclear Akt signaling.[17] P27 transcription has also been linked to another tumor suppressor gene, MEN1, in pancreatic islet cells where it promotes CDKN1B expression.[17]

Translation edit

Translation of CDKN1B reaches its maximum during quiescence and early G1.[17] Translation is regulated by polypyrimidine tract-binding protein(PTB), ELAVL1, ELAVL4, and microRNAs.[17] PTB acts by binding CDKN1b IRES to increase translation and when PTB levels decrease, G1 phase is shortened.[17] ELAVL1 and ELAVL4 also bind to CDKN1B IRES but they do so in order to decrease translation and so depletion of either results in G1 arrest.[17]

Proteolysis edit

Degradation of the p27 protein occurs as cells exit quiescence and enter G1.[17] Protein levels continue to fall rapidly as the cell continues through G1 and enters S phase. One of the most understood mechanisms for p27 proteolysis is the polyubiquitylation of p27 by the SCFSKP2 kinase associated protein 1 (Skp1) and 2 (Skp2).[17] SKP1 and Skp2 degrades p27 after it has been phosphorylated at threonine 187 (Thr187) by either activating cyclin E- or cyclin A-CDK2. Skp2 is mainly responsible for the degradation of p27 levels that continues through S phase.[18] However it is rarely expressed in early G1 where p27 levels first begin to decrease. During early G1 proteolysis of p27 is regulated by KIP1 Ubiquitylation Promoting Complex (KPC) which binds to its CDK inhibitory domain.[19] P27 also has three Cdk-inhibited tyrosines at residues 74, 88, and 89.[17] Of these, Tyr74 is of special interest because it is specific to p27-type inhibitors.[17]

Nuclear export edit

Alternatively to the transcription, translation, and proteolytic method of regulation, p27 levels can also be changed by exporting p27 to the cytoplasm. This occurs when p27 is phosphorylated on Ser(10) which allows for CRM1, a nuclear export carrier protein, to bind to and remove p27 from the nucleus.[20] Once p27 is excluded from the nucleus it cannot inhibit the cell's growth. In the cytoplasm it may be degraded entirely or retained.[16] This step occurs very early when the cell is exiting the quiescent phase and thus is independent of Skp2 degradation of p27.[20]

MicroRNA regulation edit

Because p27 levels can be moderated at the translational level, it has been proposed that p27 may be regulated by miRNAs. Recent research has suggested that both miR-221 and miR-222 control p27 levels although the pathways are not well understood.[16]

Role in cancer edit

Proliferation edit

p27 is considered a tumor suppressor because of its function as a regulator of the cell cycle.[17] In cancers it is often inactivated via impaired synthesis, accelerated degradation, or mislocalization.[17] Inactivation of p27 is generally accomplished post-transcription by the oncogenic activation of various pathways including receptor tyrosine kinases (RTK), phosphatilidylinositol 3-kinase (PI3K), SRC, or Ras-mitogen activated protein kinase(MAPK).[17] These act to accelerate the proteolysis of the p27 protein and allow the cancer cell to undergo rapid division and uncontrolled proliferation.[17] When p27 is phosphorylated by Src at tyrosine 74 or 88 it ceases to inhibit cyclinE-cdk2.[21] Src was also shown to reduce the half life of p27 meaning it is degraded faster.[21] Many epithelial cancers are known to overexpress EGFR which plays a role in the proteolysis of p27 and in Ras-driven proteolysis.[17] Non-epithelial cancers use different pathways to inactivate p27.[17] Many cancer cells also upregulate Skp2 which is known to play an active role in the proteolysis of p27[18] As a result, Skp2 is inversely related to p27 levels and directly correlates with tumor grade in many malignancies.[18]

Metastasis edit

In cancer cells, p27 can also be mislocalized to the cytoplasm in order to facilitate metastasis. The mechanisms by which it acts on motility differ between cancers. In hepatocellular carcinoma cells p27 co-localizes with actin fibers to act on GTPase Rac and induce cell migration.[22] In breast cancer cytoplasmic p27 reduced RHOA activity which increased a cell's propensity for motility.[23]

This role for p27 may indicate why cancer cells rarely fully inactivate or delete p27. By retaining p27 in some capacity it can be exported to the cytoplasm during tumorigenesis and manipulated to aid in metastasis. 70% of metastatic melanomas were shown to exhibit cytoplasmic p27, while in benign melanomas p27 remained localized to the nucleus.[24] P27 is misplaced to the cytoplasm by the MAP2K, Ras, and Akt pathways although the mechanisms are not entirely understood.[25][26][27] Additionally, phosphorylation of p27 at T198 by RSK1 has been shown to mislocalize p27 to the cytoplasm as well as inhibit the RhoA pathway.[28] Because inhibition of RhoA results in a decrease in both stress fibers and focal adhesion, cell motility is increased.[29] P27 can also be exported to the cytoplasm by oncogenic activation of the P13K pathway.[29] Thus, mislocalization of p27 to the cytoplasm in cancer cells allows them to proliferate unchecked and provides for increased motility.

In contrast to these results, p27 has also been shown to be an inhibitor of migration in sarcoma cells.[30] In these cells, p27 bound to stathmin which prevents stathmin from binding to tubulin and thus polymerization of microtubules increased and cell motility decreased.[30]

MicroRNA regulation edit

Studies of various cell lines including glioblastoma cell lines, three prostate cancer cell lines, and a breast tumor cell line showed that suppressing miR-221 and miR-22 expression resulted in p27-dependent G1 growth arrest[16] Then when p27 was knocked down, cell growth resumed indicating a strong role for miRNA regulated p27.[16] Studies in patients have demonstrated an inverse correlation between miR-221&22 and p27 protein levels. Additionally nearby healthy tissue showed high expression of the p27 protein while miR-221&22 concentrations were low.[16]

Regulation in specific cancers edit

In most cancers reduced levels of nuclear p27 are correlated with increased tumor size, increased tumor grade, and a higher propensity for metastasis. However the mechanisms by which levels of p27 are regulated vary between cancers.

Breast edit

In breast cancer, Src activation has been shown to correlate with low levels of p27[21] Breast cancers that were Estrogen receptor negative and progesterone receptor negative were more likely to display low levels of p27 and more likely to have a high tumor grade.[21] Similarly, breast cancer patients with BRCA1/2 mutations were more likely to have low levels of p27.[31]

Prostate edit

A mutation in the CDKN1B gene has been linked to an increased risk for hereditary prostate cancer in humans.[32]

Multiple Endocrine Neoplasia edit

Mutations in the CDKN1B gene has been reported in families affected by the development of primary hyperparathyroidism and pituitary adenomas, and has been classified MEN4 (multiple endocrine neoplasia, type 4). Testing for CDKN1B mutations has been recommended in patients with suspected MEN, in whom previous testing for, the more common MEN1/RET mutation, is negative.[33]

Clinical significance edit

Prognostic value edit

Several studies have demonstrated that reduced p27 levels indicate a poorer patient prognosis.[17] However, because of the dual, contrasting roles p27 plays in cancer (as an inhibitor of growth and as a mechanism for metastasis) low levels of p27 may demonstrate that a cancer is not aggressive and will remain benign.[17] In ovarian cancer, p27 negative tumors progressed in 23 months compared to 85 months in p27 positive tumors and thus could be used as a prognostic marker.[34] Similar studies have correlated low levels of p27 with a worse prognosis in breast cancer.[35] Colorectal carcinomas that lacked p27 were shown to have increased p27-specific proteolysis and a median survival of only 69 months compared to 151 months for patients with high or normal levels of p27.[36] The authors proposed clinicians could use patient specific levels of p27 to determine who would benefit from adjuvant therapy.[36] Similar correlations were observed in patients with non-small cell lung cancer,[37] those with colon,[37] and prostate cancer.[38]

So far studies have only evaluated the prognostic value of p27 retrospectively and a standardized scoring system has not been established.[17] However it has been proposed that clinicians should evaluate a patient's p27 levels in order to determine if they will be responsive to certain chemotoxins which target fast growing tumors where p27 levels are low.[17] Or in contrast, if p27 levels are found to be high in a patient's cancer, their risk for metastasis is higher and the physician can make an informed decision about their treatment plan.[17] Because p27 levels are controlled post-transcriptionally, proteomic surveys can be used to establish and monitor a patient's individual levels which aids in the future of individualized medicine.

The following cancers have been demonstrated to have an inverse correlation with p27 expression and prognosis: oro-pharyngo-laryngeal, oesophageal, gastric, colon, lung, melanoma, glioma, breast cancer, prostate, lymphoma, leukemia.[18]

Correlation to treatment response edit

P27 may also allow clinicians to better select an appropriate treatment for a patient. For example, patients with non-small cell lung cancer who were treated with platinum based chemotherapy showed reduced survival if they had low levels of p27.[39] Similarly low levels of p27 correlated with poor results from adjuvant chemotherapy in breast cancer patients.[40]

Value as a therapeutic target edit

P27 has been explored as a potential target for cancer therapy because its levels are highly correlated to patient prognosis.[41] This is true for a wide spectrum of cancers including colon, breast, prostate, lung, liver, stomach, and bladder.[41]

Use of microRNAs for therapy edit

Because of the role miRNAs play in p27 regulation, research is underway to determine if antagomiRs that block the activity of the miR221&222 and allow for p27 cell grow inhibition to take place could act as therapeutic cancer drugs.[16]

Role in Regeneration edit

Knockdown of CDKN1B stimulates regeneration of cochlear hair cells in mice. Since CDKN1B prevents cells from entering the cell cycle, inhibition of the protein could cause re-entry and subsequent division. In mammals where regeneration of cochlear hair cells normally does not occur, this inhibition could help regrow damaged cells who are otherwise incapable of proliferation. In fact, when the CDKN1B gene is disrupted in adult mice, hair cells of the organ of Corti proliferate, while those in control mice do not. Lack of CDKN1B expression appears to release the hair cells from natural cell-cycle arrest.[42][43] Because hair cell death in the human cochlea is a major cause of hearing loss, the CDKN1B protein could be an important factor in the clinical treatment of deafness.

Interactions edit

CDKN1B has been shown to interact with:

 
Overview of signal transduction pathways involved in apoptosis.

See also edit

References edit

  1. ^ a b c GRCh38: Ensembl release 89: ENSG00000111276 - Ensembl, May 2017
  2. ^ a b c GRCm38: Ensembl release 89: ENSMUSG00000003031 - 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. ^ Polyak K, Lee MH, Erdjument-Bromage H, Koff A, Roberts JM, Tempst P, Massagué J (August 1994). "Cloning of p27Kip1, a cyclin-dependent kinase inhibitor and a potential mediator of extracellular antimitogenic signals". Cell. 78 (1): 59–66. doi:10.1016/0092-8674(94)90572-X. PMID 8033212. S2CID 38513201.
  6. ^ Chiarle, R; Pagano, M; Inghirami, G (2001). "The cyclin dependent kinase inhibitor p27 and its prognostic role in breast cancer". Breast Cancer Research. 3 (2): 91–4. doi:10.1186/bcr277. PMC 139437. PMID 11250752.
  7. ^ Cooper/ Hausman, Geoffrey/ Robert (2009). The Cell. Boston University: ASM Press. p. 669. ISBN 978-0-87893-300-6.
  8. ^ Fero ML, Rivkin M, Tasch M, Porter P, Carow CE, Firpo E, Polyak K, Tsai LH, Broudy V, Perlmutter RM, Kaushansky K, Roberts JM (May 1996). "A syndrome of multiorgan hyperplasia with features of gigantism, tumorigenesis, and female sterility in p27(Kip1)-deficient mice". Cell. 85 (5): 733–44. doi:10.1016/S0092-8674(00)81239-8. PMID 8646781. S2CID 15490866.
  9. ^ Kiyokawa H, Kineman RD, Manova-Todorova KO, Soares VC, Hoffman ES, Ono M, Khanam D, Hayday AC, Frohman LA, Koff A (May 1996). "Enhanced growth of mice lacking the cyclin-dependent kinase inhibitor function of p27(Kip1)". Cell. 85 (5): 721–32. doi:10.1016/S0092-8674(00)81238-6. PMID 8646780. S2CID 19030818.
  10. ^ Nakayama K, Ishida N, Shirane M, Inomata A, Inoue T, Shishido N, Horii I, Loh DY, Nakayama K (May 1996). "Mice lacking p27(Kip1) display increased body size, multiple organ hyperplasia, retinal dysplasia, and pituitary tumors". Cell. 85 (5): 707–20. doi:10.1016/S0092-8674(00)81237-4. PMID 8646779. S2CID 2009281.
  11. ^ Klopfleisch R, Gruber AD (January 2009). "Differential expression of cell cycle regulators p21, p27 and p53 in metastasizing canine mammary adenocarcinomas versus normal mammary glands". Res Vet Sci. 87 (1): 91–6. doi:10.1016/j.rvsc.2008.12.010. PMID 19185891.
  12. ^ Klopfleisch R, Schütze M, Gruber AD (September 2010). "Loss of p27 expression in canine mammary tumors and their metastases". Res Vet Sci. 88 (2): 300–3. doi:10.1016/j.rvsc.2009.08.007. PMID 19748645.
  13. ^ Klopfleisch R, von Euler H, Sarli G, Pinho SS, Gärtner F, Gruber AD (2010). "Molecular Carcinogenesis of Canine Mammary Tumors: News From an Old Disease". Veterinary Pathology. 48 (1): 98–116. doi:10.1177/0300985810390826. PMID 21149845. S2CID 206509356.
  14. ^ Klopfleisch R, Schütze M, Gruber AD (October 2009). "Downregulation of transforming growth factor β (TGFβ) and latent TGFβ binding protein (LTBP)-4 expression in late stage canine mammary tumours". Veterinary Journal. 186 (3): 379–84. doi:10.1016/j.tvjl.2009.09.014. PMID 19836277.
  15. ^ Göpfert U, Kullmann M, Hengst L (July 2003). "Cell cycle-dependent translation of p27 involves a responsive element in its 5'-UTR that overlaps with a uORF". Hum. Mol. Genet. 12 (14): 1767–79. doi:10.1093/hmg/ddg177. PMID 12837699.
  16. ^ a b c d e f g le Sage C, Nagel R, Agami R (November 2007). "Diverse ways to control p27Kip1 function: miRNAs come into play". Cell Cycle. 6 (22): 2742–9. doi:10.4161/cc.6.22.4900. PMID 17986865.
  17. ^ a b c d e f g h i j k l m n o p q r s t u v Chu IM, Hengst L, Slingerland JM (April 2008). "The Cdk inhibitor p27 in human cancer: prognostic potential and relevance to anticancer therapy". Nat. Rev. Cancer. 8 (4): 253–67. doi:10.1038/nrc2347. PMID 18354415. S2CID 2175257.
  18. ^ a b c d Nakayama KI, Nakayama K (May 2006). "Ubiquitin ligases: cell-cycle control and cancer". Nat. Rev. Cancer. 6 (5): 369–81. doi:10.1038/nrc1881. PMID 16633365. S2CID 19594293.
  19. ^ Kotoshiba S, Kamura T, Hara T, Ishida N, Nakayama KI (May 2005). "Molecular dissection of the interaction between p27 and Kip1 ubiquitylation-promoting complex, the ubiquitin ligase that regulates proteolysis of p27 in G1 phase". J. Biol. Chem. 280 (18): 17694–700. doi:10.1074/jbc.M500866200. PMID 15746103.
  20. ^ a b Ishida N, Hara T, Kamura T, Yoshida M, Nakayama K, Nakayama KI (April 2002). "Phosphorylation of p27Kip1 on serine 10 is required for its binding to CRM1 and nuclear export". J. Biol. Chem. 277 (17): 14355–8. doi:10.1074/jbc.C100762200. PMID 11889117.
  21. ^ a b c d Chu I, Sun J, Arnaout A, Kahn H, Hanna W, Narod S, Sun P, Tan CK, Hengst L, Slingerland J (January 2007). "p27 phosphorylation by Src regulates inhibition of cyclin E-Cdk2". Cell. 128 (2): 281–94. doi:10.1016/j.cell.2006.11.049. PMC 1961623. PMID 17254967.
  22. ^ McAllister SS, Becker-Hapak M, Pintucci G, Pagano M, Dowdy SF (January 2003). "Novel p27(kip1) C-terminal scatter domain mediates Rac-dependent cell migration independent of cell cycle arrest functions". Mol. Cell. Biol. 23 (1): 216–28. doi:10.1128/MCB.23.1.216-228.2003. PMC 140659. PMID 12482975.
  23. ^ Wu FY, Wang SE, Sanders ME, Shin I, Rojo F, Baselga J, Arteaga CL (February 2006). "Reduction of cytosolic p27(Kip1) inhibits cancer cell motility, survival, and tumorigenicity". Cancer Res. 66 (4): 2162–72. doi:10.1158/0008-5472.CAN-05-3304. PMID 16489017.
  24. ^ Denicourt C, Saenz CC, Datnow B, Cui XS, Dowdy SF (October 2007). "Relocalized p27Kip1 tumor suppressor functions as a cytoplasmic metastatic oncogene in melanoma". Cancer Res. 67 (19): 9238–43. doi:10.1158/0008-5472.CAN-07-1375. PMID 17909030.
  25. ^ Cheng M, Sexl V, Sherr CJ, Roussel MF (February 1998). "Assembly of cyclin D-dependent kinase and titration of p27Kip1 regulated by mitogen-activated protein kinase kinase (MEK1)". Proc. Natl. Acad. Sci. U.S.A. 95 (3): 1091–6. Bibcode:1998PNAS...95.1091C. doi:10.1073/pnas.95.3.1091. PMC 18683. PMID 9448290.
  26. ^ Liu X, Sun Y, Ehrlich M, Lu T, Kloog Y, Weinberg RA, Lodish HF, Henis YI (November 2000). "Disruption of TGF-beta growth inhibition by oncogenic ras is linked to p27Kip1 mislocalization". Oncogene. 19 (51): 5926–35. doi:10.1038/sj.onc.1203991. PMID 11127824.
  27. ^ Viglietto G, Motti ML, Bruni P, Melillo RM, D'Alessio A, Califano D, Vinci F, Chiappetta G, Tsichlis P, Bellacosa A, Fusco A, Santoro M (October 2002). "Cytoplasmic relocalization and inhibition of the cyclin-dependent kinase inhibitor p27(Kip1) by PKB/Akt-mediated phosphorylation in breast cancer". Nat. Med. 8 (10): 1136–44. doi:10.1038/nm762. PMID 12244303. S2CID 6580033.
  28. ^ Larrea MD, Hong F, Wander SA, da Silva TG, Helfman D, Lannigan D, Smith JA, Slingerland JM (June 2009). "RSK1 drives p27Kip1 phosphorylation at T198 to promote RhoA inhibition and increase cell motility". Proc. Natl. Acad. Sci. U.S.A. 106 (23): 9268–73. Bibcode:2009PNAS..106.9268L. doi:10.1073/pnas.0805057106. PMC 2695095. PMID 19470470.
  29. ^ a b Larrea MD, Wander SA, Slingerland JM (November 2009). "p27 as Jekyll and Hyde: regulation of cell cycle and cell motility". Cell Cycle. 8 (21): 3455–61. doi:10.4161/cc.8.21.9789. PMID 19829074.
  30. ^ a b Baldassarre G, Belletti B, Nicoloso MS, Schiappacassi M, Vecchione A, Spessotto P, Morrione A, Canzonieri V, Colombatti A (January 2005). "p27(Kip1)-stathmin interaction influences sarcoma cell migration and invasion". Cancer Cell. 7 (1): 51–63. doi:10.1016/j.ccr.2004.11.025. PMID 15652749.
  31. ^ Chappuis PO, Kapusta L, Bégin LR, Wong N, Brunet JS, Narod SA, Slingerland J, Foulkes WD (December 2000). "Germline BRCA1/2 mutations and p27(Kip1) protein levels independently predict outcome after breast cancer". J. Clin. Oncol. 18 (24): 4045–52. doi:10.1200/jco.2000.18.24.4045. PMID 11118465.
  32. ^ Chang BL, Zheng SL, Isaacs SD, Wiley KE, Turner A, Li G, Walsh PC, Meyers DA, Isaacs WB, Xu J (March 2004). "A polymorphism in the CDKN1B gene is associated with increased risk of hereditary prostate cancer". Cancer Res. 64 (6): 1997–9. doi:10.1158/0008-5472.CAN-03-2340. PMID 15026335.
  33. ^ Alrezk R, Hannah-Shmouni F, Stratakis CA (August 2017). "MEN4 and CDKN1B mutations: the latest of the MEN syndromes". Endocrine-Related Cancer. 24 (10): T195–208. doi:10.1530/ERC-17-0243. PMC 5623937. PMID 28824003.
  34. ^ Masciullo V, Sgambato A, Pacilio C, Pucci B, Ferrandina G, Palazzo J, Carbone A, Cittadini A, Mancuso S, Scambia G, Giordano A (August 1999). "Frequent loss of expression of the cyclin-dependent kinase inhibitor p27 in epithelial ovarian cancer". Cancer Res. 59 (15): 3790–4. PMID 10446997.
  35. ^ Catzavelos C, Bhattacharya N, Ung YC, Wilson JA, Roncari L, Sandhu C, Shaw P, Yeger H, Morava-Protzner I, Kapusta L, Franssen E, Pritchard KI, Slingerland JM (February 1997). "Decreased levels of the cell-cycle inhibitor p27Kip1 protein: prognostic implications in primary breast cancer". Nat. Med. 3 (2): 227–30. doi:10.1038/nm0297-227. PMID 9018244. S2CID 25460889.
  36. ^ a b Loda M, Cukor B, Tam SW, Lavin P, Fiorentino M, Draetta GF, Jessup JM, Pagano M (February 1997). "Increased proteasome-dependent degradation of the cyclin-dependent kinase inhibitor p27 in aggressive colorectal carcinomas". Nat. Med. 3 (2): 231–4. doi:10.1038/nm0297-231. PMID 9018245. S2CID 3164478.
  37. ^ a b Esposito V, Baldi A, De Luca A, Groger AM, Loda M, Giordano GG, Caputi M, Baldi F, Pagano M, Giordano A (August 1997). "Prognostic role of the cyclin-dependent kinase inhibitor p27 in non-small cell lung cancer". Cancer Res. 57 (16): 3381–5. PMID 9270000.
  38. ^ Tsihlias J, Kapusta LR, DeBoer G, Morava-Protzner I, Zbieranowski I, Bhattacharya N, Catzavelos GC, Klotz LH, Slingerland JM (February 1998). "Loss of cyclin-dependent kinase inhibitor p27Kip1 is a novel prognostic factor in localized human prostate adenocarcinoma". Cancer Res. 58 (3): 542–8. PMID 9458103.
  39. ^ Oshita F, Kameda Y, Nishio K, Tanaka G, Yamada K, Nomura I, Nakayama H, Noda K (2000). "Increased expression levels of cyclin-dependent kinase inhibitor p27 correlate with good responses to platinum-based chemotherapy in non-small cell lung cancer". Oncol. Rep. 7 (3): 491–5. doi:10.3892/or.7.3.491. PMID 10767357.
  40. ^ Porter PL, Barlow WE, Yeh IT, Lin MG, Yuan XP, Donato E, Sledge GW, Shapiro CL, Ingle JN, Haskell CM, Albain KS, Roberts JM, Livingston RB, Hayes DF (December 2006). "p27(Kip1) and cyclin E expression and breast cancer survival after treatment with adjuvant chemotherapy". J. Natl. Cancer Inst. 98 (23): 1723–31. doi:10.1093/jnci/djj467. PMC 2727647. PMID 17148774.
  41. ^ a b Blain SW, Scher HI, Cordon-Cardo C, Koff A (February 2003). "p27 as a target for cancer therapeutics". Cancer Cell. 3 (2): 111–5. doi:10.1016/S1535-6108(03)00026-6. PMID 12620406.
  42. ^ Löwenheim H, Furness DN, Kil J, Zinn C, Gültig K, Fero ML, Frost D, Gummer AW, Roberts JM, Rubel EW, Hackney CM, Zenner HP (1999). "Gene disruption of p27(Kip1) allows cell proliferation in the postnatal and adult organ of corti". Proc. Natl. Acad. Sci. U.S.A. 96 (7): 4084–8. Bibcode:1999PNAS...96.4084L. doi:10.1073/pnas.96.7.4084. PMC 22424. PMID 10097167.
  43. ^ Nakagawa T (2014). "Strategies for developing novel therapeutics for sensorineural hearing loss". Front Pharmacol. 5: 206. doi:10.3389/fphar.2014.00206. PMC 4165348. PMID 25278894.
  44. ^ a b Fujita N, Sato S, Katayama K, Tsuruo T (2002). "Akt-dependent phosphorylation of p27Kip1 promotes binding to 14-3-3 and cytoplasmic localization". J. Biol. Chem. 277 (32): 28706–13. doi:10.1074/jbc.M203668200. PMID 12042314.
  45. ^ a b Wang W, Ungermannova D, Chen L, Liu X (2003). "A negatively charged amino acid in Skp2 is required for Skp2-Cks1 interaction and ubiquitination of p27Kip1". J. Biol. Chem. 278 (34): 32390–6. doi:10.1074/jbc.M305241200. PMID 12813041.
  46. ^ a b Sitry D, Seeliger MA, Ko TK, Ganoth D, Breward SE, Itzhaki LS, Pagano M, Hershko A (2002). "Three different binding sites of Cks1 are required for p27-ubiquitin ligation". J. Biol. Chem. 277 (44): 42233–40. doi:10.1074/jbc.M205254200. PMID 12140288.
  47. ^ a b Lin J, Jinno S, Okayama H (2001). "Cdk6-cyclin D3 complex evades inhibition by inhibitor proteins and uniquely controls cell's proliferation competence". Oncogene. 20 (16): 2000–9. doi:10.1038/sj.onc.1204375. PMID 11360184.
  48. ^ Rual JF, Venkatesan K, Hao T, Hirozane-Kishikawa T, Dricot A, Li N, Berriz GF, Gibbons FD, Dreze M, Ayivi-Guedehoussou N, Klitgord N, Simon C, Boxem M, Milstein S, Rosenberg J, Goldberg DS, Zhang LV, Wong SL, Franklin G, Li S, Albala JS, Lim J, Fraughton C, Llamosas E, Cevik S, Bex C, Lamesch P, Sikorski RS, Vandenhaute J, Zoghbi HY, Smolyar A, Bosak S, Sequerra R, Doucette-Stamm L, Cusick ME, Hill DE, Roth FP, Vidal M (2005). "Towards a proteome-scale map of the human protein-protein interaction network". Nature. 437 (7062): 1173–8. Bibcode:2005Natur.437.1173R. doi:10.1038/nature04209. PMID 16189514. S2CID 4427026.
  49. ^ Zhang Q, Wang X, Wolgemuth DJ (1999). "Developmentally regulated expression of cyclin D3 and its potential in vivo interacting proteins during murine gametogenesis". Endocrinology. 140 (6): 2790–800. doi:10.1210/endo.140.6.6756. PMID 10342870.
  50. ^ a b c Connor MK, Kotchetkov R, Cariou S, Resch A, Lupetti R, Beniston RG, Melchior F, Hengst L, Slingerland JM (2003). "CRM1/Ran-mediated nuclear export of p27(Kip1) involves a nuclear export signal and links p27 export and proteolysis". Mol. Biol. Cell. 14 (1): 201–13. doi:10.1091/mbc.E02-06-0319. PMC 140238. PMID 12529437.
  51. ^ Shanahan F, Seghezzi W, Parry D, Mahony D, Lees E (1999). "Cyclin E associates with BAF155 and BRG1, components of the mammalian SWI-SNF complex, and alters the ability of BRG1 to induce growth arrest". Mol. Cell. Biol. 19 (2): 1460–9. doi:10.1128/mcb.19.2.1460. PMC 116074. PMID 9891079.
  52. ^ a b Porter LA, Kong-Beltran M, Donoghue DJ (2003). "Spy1 interacts with p27Kip1 to allow G1/S progression". Mol. Biol. Cell. 14 (9): 3664–74. doi:10.1091/mbc.E02-12-0820. PMC 196558. PMID 12972555.
  53. ^ Youn CK, Cho HJ, Kim SH, Kim HB, Kim MH, Chang IY, Lee JS, Chung MH, Hahm KS, You HJ (2005). "Bcl-2 expression suppresses mismatch repair activity through inhibition of E2F transcriptional activity". Nat. Cell Biol. 7 (2): 137–47. doi:10.1038/ncb1215. PMID 15619620. S2CID 42766325.
  54. ^ Law BK, Chytil A, Dumont N, Hamilton EG, Waltner-Law ME, Aakre ME, Covington C, Moses HL (2002). "Rapamycin potentiates transforming growth factor beta-induced growth arrest in nontransformed, oncogene-transformed, and human cancer cells". Mol. Cell. Biol. 22 (23): 8184–98. doi:10.1128/MCB.22.23.8184-8198.2002. PMC 134072. PMID 12417722.
  55. ^ Rosner M, Hengstschläger M (2004). "Tuberin binds p27 and negatively regulates its interaction with the SCF component Skp2". J. Biol. Chem. 279 (47): 48707–15. doi:10.1074/jbc.M405528200. PMID 15355997.
  56. ^ Cariou S, Donovan JC, Flanagan WM, Milic A, Bhattacharya N, Slingerland JM (2000). "Down-regulation of p21WAF1/CIP1 or p27Kip1 abrogates antiestrogen-mediated cell cycle arrest in human breast cancer cells". Proc. Natl. Acad. Sci. U.S.A. 97 (16): 9042–6. Bibcode:2000PNAS...97.9042C. doi:10.1073/pnas.160016897. PMC 16818. PMID 10908655.
  57. ^ Sugiyama Y, Tomoda K, Tanaka T, Arata Y, Yoneda-Kato N, Kato J (2001). "Direct binding of the signal-transducing adaptor Grb2 facilitates down-regulation of the cyclin-dependent kinase inhibitor p27Kip1". J. Biol. Chem. 276 (15): 12084–90. doi:10.1074/jbc.M010811200. PMID 11278754.
  58. ^ Smitherman M, Lee K, Swanger J, Kapur R, Clurman BE (2000). "Characterization and targeted disruption of murine Nup50, a p27(Kip1)-interacting component of the nuclear pore complex". Mol. Cell. Biol. 20 (15): 5631–42. doi:10.1128/MCB.20.15.5631-5642.2000. PMC 86029. PMID 10891500.
  59. ^ Ishida N, Hara T, Kamura T, Yoshida M, Nakayama K, Nakayama KI (2002). "Phosphorylation of p27Kip1 on serine 10 is required for its binding to CRM1 and nuclear export". J. Biol. Chem. 277 (17): 14355–8. doi:10.1074/jbc.C100762200. PMID 11889117.

Further reading edit

  • Marone M, Bonanno G, Rutella S, Leone G, Scambia G, Pierelli L (2003). "Survival and cell cycle control in early hematopoiesis: role of bcl-2, and the cyclin dependent kinase inhibitors P27 and P21". Leuk. Lymphoma. 43 (1): 51–7. doi:10.1080/10428190210195. PMID 11908736. S2CID 28490341.
  • Hirabayashi H (2003). "[P27 expression and survival in NSCLC]". Nippon Rinsho. 60 Suppl 5: 263–6. PMID 12101669.
  • Bloom J, Pagano M (2003). "Deregulated degradation of the cdk inhibitor p27 and malignant transformation". Semin. Cancer Biol. 13 (1): 41–7. CiteSeerX 10.1.1.513.177. doi:10.1016/S1044-579X(02)00098-6. PMID 12507555.
  • Tokumoto M, Tsuruya K, Fukuda K, Kanai H, Kuroki S, Hirakata H, Iida M (2003). "Parathyroid cell growth in patients with advanced secondary hyperparathyroidism: vitamin D receptor and cyclin-dependent kinase inhibitors, p21 and p27". Nephrol. Dial. Transplant. 18 Suppl 3 (90003): iii9–12. doi:10.1093/ndt/gfg1003. PMID 12771291.
  • Drexler HC (2004). "The role of p27Kip1 in proteasome inhibitor induced apoptosis". Cell Cycle. 2 (5): 438–41. doi:10.4161/cc.2.5.461. PMID 12963837.
  • Le XF, Pruefer F, Bast RC (2006). "HER2-targeting antibodies modulate the cyclin-dependent kinase inhibitor p27Kip1 via multiple signaling pathways". Cell Cycle. 4 (1): 87–95. doi:10.4161/cc.4.1.1360. PMID 15611642.
  • Belletti B, Nicoloso MS, Schiappacassi M, Chimienti E, Berton S, Lovat F, Colombatti A, Baldassarre G (2005). "p27(kip1) functional regulation in human cancer: a potential target for therapeutic designs". Curr. Med. Chem. 12 (14): 1589–605. doi:10.2174/0929867054367149. PMID 16022660.
  • Sankaranarayanan P, Schomay TE, Aiello KA, Alter O (April 2015). "Tensor GSVD of Patient- and Platform-Matched Tumor and Normal DNA Copy-Number Profiles Uncovers Chromosome Arm-Wide Patterns of Tumor-Exclusive Platform-Consistent Alterations Encoding for Cell Transformation and Predicting Ovarian Cancer Survival". PLOS ONE. 10 (4): e0121396. Bibcode:2015PLoSO..1021396S. doi:10.1371/journal.pone.0121396. PMC 4398562. PMID 25875127.

External links edit

cdkn1b, cyclin, dependent, kinase, inhibitor, p27kip1, enzyme, inhibitor, that, humans, encoded, gene, encodes, protein, which, belongs, family, cyclin, dependent, kinase, inhibitor, proteins, encoded, protein, binds, prevents, activation, cyclin, cdk2, cyclin. Cyclin dependent kinase inhibitor 1B p27Kip1 is an enzyme inhibitor that in humans is encoded by the CDKN1B gene 5 It encodes a protein which belongs to the Cip Kip family of cyclin dependent kinase Cdk inhibitor proteins The encoded protein binds to and prevents the activation of cyclin E CDK2 or cyclin D CDK4 complexes and thus controls the cell cycle progression at G1 It is often referred to as a cell cycle inhibitor protein because its major function is to stop or slow down the cell division cycle CDKN1BAvailable structuresPDBOrtholog search PDBe RCSBList of PDB id codes2AST 1H27 1JSUIdentifiersAliasesCDKN1B CDKN4 KIP1 MEN1B MEN4 P27KIP1 cyclin dependent kinase inhibitor 1B cyclin dependent kinase inhibitor 1BExternal IDsOMIM 600778 MGI 104565 HomoloGene 2999 GeneCards CDKN1BGene location Human Chr Chromosome 12 human 1 Band12p13 1Start12 685 498 bp 1 End12 722 369 bp 1 Gene location Mouse Chr Chromosome 6 mouse 2 Band6 G1 6 65 77 cMStart134 897 364 bp 2 End134 902 476 bp 2 RNA expression patternBgeeHumanMouse ortholog Top expressed inretinal pigment epitheliumganglionic eminenceponsspongy boneparietal pleurasuperficial temporal arterygerminal epitheliumcerebellar vermissubthalamic nucleusendothelial cellTop expressed incerebellar vermisbloodpineal glandcumulus cellhabenulaganglionic eminenceseminal vesiculamedian eminencedorsomedial hypothalamic nucleusaortic valveMore reference expression dataBioGPSMore reference expression dataGene ontologyMolecular functionprotein binding protein phosphatase binding cyclin dependent protein serine threonine kinase activity protein kinase inhibitor activity protein kinase binding cyclin binding cyclin dependent protein serine threonine kinase inhibitor activity Hsp70 protein binding protein containing complex binding chaperone bindingCellular componentcytoplasm cytosol endosome nucleoplasm Cul4A RING E3 ubiquitin ligase complex nucleus intracellular membrane bounded organelle protein containing complexBiological processNotch signaling pathway regulation of cyclin dependent protein serine threonine kinase activity DNA damage response signal transduction by p53 class mediator resulting in cell cycle arrest response to estradiol positive regulation of cell death response to hypoxia response to amino acid response to cadmium ion positive regulation of protein catabolic process response to organic cyclic compound cellular response to lithium ion cellular response to organic cyclic compound response to peptide hormone cell death negative regulation of apoptotic process sensory perception of sound response to glucose potassium ion transport regulation of cell population proliferation negative regulation of cell growth negative regulation of epithelial cell proliferation involved in prostate gland development positive regulation of cell population proliferation positive regulation of cyclin dependent protein serine threonine kinase activity negative regulation of epithelial cell proliferation positive regulation of microtubule polymerization inner ear development cellular response to antibiotic cell cycle negative regulation of phosphorylation negative regulation of transcription DNA templated autophagic cell death negative regulation of cell population proliferation negative regulation of mitotic cell cycle negative regulation of kinase activity positive regulation of cell cycle negative regulation of vascular associated smooth muscle cell proliferation placenta development regulation of exit from mitosis regulation of lens fiber cell differentiation negative regulation of cyclin dependent protein kinase activity negative regulation of cell cycle negative regulation of cyclin dependent protein serine threonine kinase activity G1 S transition of mitotic cell cycleSources Amigo QuickGOOrthologsSpeciesHumanMouseEntrez102712576EnsemblENSG00000111276ENSMUSG00000003031UniProtP46527P46414RefSeq mRNA NM 004064NM 009875RefSeq protein NP 004055NP 034005Location UCSC Chr 12 12 69 12 72 MbChr 6 134 9 134 9 MbPubMed search 3 4 WikidataView Edit HumanView Edit Mouse Contents 1 Function 2 Regulation 2 1 Transcription 2 2 Translation 2 3 Proteolysis 2 4 Nuclear export 2 5 MicroRNA regulation 3 Role in cancer 3 1 Proliferation 3 2 Metastasis 3 3 MicroRNA regulation 4 Regulation in specific cancers 4 1 Breast 4 2 Prostate 4 3 Multiple Endocrine Neoplasia 5 Clinical significance 5 1 Prognostic value 5 2 Correlation to treatment response 5 3 Value as a therapeutic target 5 3 1 Use of microRNAs for therapy 6 Role in Regeneration 7 Interactions 8 See also 9 References 10 Further reading 11 External linksFunction editThe p27Kip1 gene has a DNA sequence similar to other members of the Cip Kip family which include the p21Cip1 Waf1 and p57Kip2 genes In addition to this structural similarity the Cip Kip proteins share the functional characteristic of being able to bind several different classes of Cyclin and Cdk molecules For example p27Kip1 binds to cyclin D either alone or when complexed to its catalytic subunit CDK4 In doing so p27Kip1 inhibits the catalytic activity of Cdk4 which means that it prevents Cdk4 from adding phosphate residues to its principal substrate the retinoblastoma pRb protein Increased levels of the p27Kip1 protein typically cause cells to arrest in the G1 phase of the cell cycle Likewise p27Kip1 is able to bind other Cdk proteins when complexed to cyclin subunits such as Cyclin E Cdk2 and Cyclin A Cdk2 6 Regulation editIn general extracellular growth factors which promote cell division reduce transcription and translation of p27Kip1 Also increased synthesis of CDk4 6 cyclin D causes binding of p27 to this complex sequestering it from binding to the CDk2 cyclin E complex Furthermore an active CDK2 cyclin E complex will phosphorylate p27 and tag p27 for ubiquitination 7 A mutation of this gene may lead to loss of control over the cell cycle leading to uncontrolled cellular proliferation 8 9 10 Loss of p27 expression has been observed in metastatic canine mammary carcinomas 11 12 13 Decreased TGF beta signalling has been suggested to cause loss of p27 expression in this tumor type 14 A structured cis regulatory element has been found in the 5 UTR of the P27 mRNA where it is thought to regulate translation relative to cell cycle progression 15 P27 regulation is accomplished by two different mechanisms In the first its concentration is changed by the individual rates of transcription translation and proteolysis P27 can also be regulated by changing its subcellular location 16 Both mechanisms act to reduce levels of p27 allowing for the activation of Cdk1 and Cdk2 and for the cell to begin progressing through the cell cycle Transcription edit Transcription of the CDKN1B gene is activated by Forkhead box class O family FoxO proteins which also acts downstream to promote p27 nuclear localization and decrease levels of COP9 subunit 5 COPS5 which helps in the degradation of p27 17 Transcription for p27 is activated by FoxO in response to cytokines promyelocytic leukaemia proteins and nuclear Akt signaling 17 P27 transcription has also been linked to another tumor suppressor gene MEN1 in pancreatic islet cells where it promotes CDKN1B expression 17 Translation edit Translation of CDKN1B reaches its maximum during quiescence and early G1 17 Translation is regulated by polypyrimidine tract binding protein PTB ELAVL1 ELAVL4 and microRNAs 17 PTB acts by binding CDKN1b IRES to increase translation and when PTB levels decrease G1 phase is shortened 17 ELAVL1 and ELAVL4 also bind to CDKN1B IRES but they do so in order to decrease translation and so depletion of either results in G1 arrest 17 Proteolysis edit Degradation of the p27 protein occurs as cells exit quiescence and enter G1 17 Protein levels continue to fall rapidly as the cell continues through G1 and enters S phase One of the most understood mechanisms for p27 proteolysis is the polyubiquitylation of p27 by the SCFSKP2 kinase associated protein 1 Skp1 and 2 Skp2 17 SKP1 and Skp2 degrades p27 after it has been phosphorylated at threonine 187 Thr187 by either activating cyclin E or cyclin A CDK2 Skp2 is mainly responsible for the degradation of p27 levels that continues through S phase 18 However it is rarely expressed in early G1 where p27 levels first begin to decrease During early G1 proteolysis of p27 is regulated by KIP1 Ubiquitylation Promoting Complex KPC which binds to its CDK inhibitory domain 19 P27 also has three Cdk inhibited tyrosines at residues 74 88 and 89 17 Of these Tyr74 is of special interest because it is specific to p27 type inhibitors 17 Nuclear export edit Alternatively to the transcription translation and proteolytic method of regulation p27 levels can also be changed by exporting p27 to the cytoplasm This occurs when p27 is phosphorylated on Ser 10 which allows for CRM1 a nuclear export carrier protein to bind to and remove p27 from the nucleus 20 Once p27 is excluded from the nucleus it cannot inhibit the cell s growth In the cytoplasm it may be degraded entirely or retained 16 This step occurs very early when the cell is exiting the quiescent phase and thus is independent of Skp2 degradation of p27 20 MicroRNA regulation edit Because p27 levels can be moderated at the translational level it has been proposed that p27 may be regulated by miRNAs Recent research has suggested that both miR 221 and miR 222 control p27 levels although the pathways are not well understood 16 Role in cancer editProliferation edit p27 is considered a tumor suppressor because of its function as a regulator of the cell cycle 17 In cancers it is often inactivated via impaired synthesis accelerated degradation or mislocalization 17 Inactivation of p27 is generally accomplished post transcription by the oncogenic activation of various pathways including receptor tyrosine kinases RTK phosphatilidylinositol 3 kinase PI3K SRC or Ras mitogen activated protein kinase MAPK 17 These act to accelerate the proteolysis of the p27 protein and allow the cancer cell to undergo rapid division and uncontrolled proliferation 17 When p27 is phosphorylated by Src at tyrosine 74 or 88 it ceases to inhibit cyclinE cdk2 21 Src was also shown to reduce the half life of p27 meaning it is degraded faster 21 Many epithelial cancers are known to overexpress EGFR which plays a role in the proteolysis of p27 and in Ras driven proteolysis 17 Non epithelial cancers use different pathways to inactivate p27 17 Many cancer cells also upregulate Skp2 which is known to play an active role in the proteolysis of p27 18 As a result Skp2 is inversely related to p27 levels and directly correlates with tumor grade in many malignancies 18 Metastasis edit In cancer cells p27 can also be mislocalized to the cytoplasm in order to facilitate metastasis The mechanisms by which it acts on motility differ between cancers In hepatocellular carcinoma cells p27 co localizes with actin fibers to act on GTPase Rac and induce cell migration 22 In breast cancer cytoplasmic p27 reduced RHOA activity which increased a cell s propensity for motility 23 This role for p27 may indicate why cancer cells rarely fully inactivate or delete p27 By retaining p27 in some capacity it can be exported to the cytoplasm during tumorigenesis and manipulated to aid in metastasis 70 of metastatic melanomas were shown to exhibit cytoplasmic p27 while in benign melanomas p27 remained localized to the nucleus 24 P27 is misplaced to the cytoplasm by the MAP2K Ras and Akt pathways although the mechanisms are not entirely understood 25 26 27 Additionally phosphorylation of p27 at T198 by RSK1 has been shown to mislocalize p27 to the cytoplasm as well as inhibit the RhoA pathway 28 Because inhibition of RhoA results in a decrease in both stress fibers and focal adhesion cell motility is increased 29 P27 can also be exported to the cytoplasm by oncogenic activation of the P13K pathway 29 Thus mislocalization of p27 to the cytoplasm in cancer cells allows them to proliferate unchecked and provides for increased motility In contrast to these results p27 has also been shown to be an inhibitor of migration in sarcoma cells 30 In these cells p27 bound to stathmin which prevents stathmin from binding to tubulin and thus polymerization of microtubules increased and cell motility decreased 30 MicroRNA regulation edit Studies of various cell lines including glioblastoma cell lines three prostate cancer cell lines and a breast tumor cell line showed that suppressing miR 221 and miR 22 expression resulted in p27 dependent G1 growth arrest 16 Then when p27 was knocked down cell growth resumed indicating a strong role for miRNA regulated p27 16 Studies in patients have demonstrated an inverse correlation between miR 221 amp 22 and p27 protein levels Additionally nearby healthy tissue showed high expression of the p27 protein while miR 221 amp 22 concentrations were low 16 Regulation in specific cancers editIn most cancers reduced levels of nuclear p27 are correlated with increased tumor size increased tumor grade and a higher propensity for metastasis However the mechanisms by which levels of p27 are regulated vary between cancers Breast edit In breast cancer Src activation has been shown to correlate with low levels of p27 21 Breast cancers that were Estrogen receptor negative and progesterone receptor negative were more likely to display low levels of p27 and more likely to have a high tumor grade 21 Similarly breast cancer patients with BRCA1 2 mutations were more likely to have low levels of p27 31 Prostate edit A mutation in the CDKN1B gene has been linked to an increased risk for hereditary prostate cancer in humans 32 Multiple Endocrine Neoplasia edit Mutations in the CDKN1B gene has been reported in families affected by the development of primary hyperparathyroidism and pituitary adenomas and has been classified MEN4 multiple endocrine neoplasia type 4 Testing for CDKN1B mutations has been recommended in patients with suspected MEN in whom previous testing for the more common MEN1 RET mutation is negative 33 Clinical significance editPrognostic value edit Several studies have demonstrated that reduced p27 levels indicate a poorer patient prognosis 17 However because of the dual contrasting roles p27 plays in cancer as an inhibitor of growth and as a mechanism for metastasis low levels of p27 may demonstrate that a cancer is not aggressive and will remain benign 17 In ovarian cancer p27 negative tumors progressed in 23 months compared to 85 months in p27 positive tumors and thus could be used as a prognostic marker 34 Similar studies have correlated low levels of p27 with a worse prognosis in breast cancer 35 Colorectal carcinomas that lacked p27 were shown to have increased p27 specific proteolysis and a median survival of only 69 months compared to 151 months for patients with high or normal levels of p27 36 The authors proposed clinicians could use patient specific levels of p27 to determine who would benefit from adjuvant therapy 36 Similar correlations were observed in patients with non small cell lung cancer 37 those with colon 37 and prostate cancer 38 So far studies have only evaluated the prognostic value of p27 retrospectively and a standardized scoring system has not been established 17 However it has been proposed that clinicians should evaluate a patient s p27 levels in order to determine if they will be responsive to certain chemotoxins which target fast growing tumors where p27 levels are low 17 Or in contrast if p27 levels are found to be high in a patient s cancer their risk for metastasis is higher and the physician can make an informed decision about their treatment plan 17 Because p27 levels are controlled post transcriptionally proteomic surveys can be used to establish and monitor a patient s individual levels which aids in the future of individualized medicine The following cancers have been demonstrated to have an inverse correlation with p27 expression and prognosis oro pharyngo laryngeal oesophageal gastric colon lung melanoma glioma breast cancer prostate lymphoma leukemia 18 Correlation to treatment response edit P27 may also allow clinicians to better select an appropriate treatment for a patient For example patients with non small cell lung cancer who were treated with platinum based chemotherapy showed reduced survival if they had low levels of p27 39 Similarly low levels of p27 correlated with poor results from adjuvant chemotherapy in breast cancer patients 40 Value as a therapeutic target edit P27 has been explored as a potential target for cancer therapy because its levels are highly correlated to patient prognosis 41 This is true for a wide spectrum of cancers including colon breast prostate lung liver stomach and bladder 41 Use of microRNAs for therapy edit Because of the role miRNAs play in p27 regulation research is underway to determine if antagomiRs that block the activity of the miR221 amp 222 and allow for p27 cell grow inhibition to take place could act as therapeutic cancer drugs 16 Role in Regeneration editKnockdown of CDKN1B stimulates regeneration of cochlear hair cells in mice Since CDKN1B prevents cells from entering the cell cycle inhibition of the protein could cause re entry and subsequent division In mammals where regeneration of cochlear hair cells normally does not occur this inhibition could help regrow damaged cells who are otherwise incapable of proliferation In fact when the CDKN1B gene is disrupted in adult mice hair cells of the organ of Corti proliferate while those in control mice do not Lack of CDKN1B expression appears to release the hair cells from natural cell cycle arrest 42 43 Because hair cell death in the human cochlea is a major cause of hearing loss the CDKN1B protein could be an important factor in the clinical treatment of deafness Interactions editCDKN1B has been shown to interact with AKT1 44 CKS1B 45 46 Cyclin D3 47 48 49 Cyclin E1 50 51 Cyclin dependent kinase 2 50 52 53 54 55 Cyclin dependent kinase 4 47 56 Grb2 57 NUP50 58 SKP2 44 45 46 SPDYA 52 and XPO1 50 59 nbsp Overview of signal transduction pathways involved in apoptosis See also editSic1 homologue in Saccharomyces cerevisiae P21waf 1 another CDK inhibitor Hyaluronic acid synthase HyaluronidaseReferences edit a b c GRCh38 Ensembl release 89 ENSG00000111276 Ensembl May 2017 a b c GRCm38 Ensembl release 89 ENSMUSG00000003031 Ensembl May 2017 Human PubMed Reference National Center for Biotechnology Information U S National Library of Medicine Mouse PubMed Reference National Center for Biotechnology Information U S National Library of Medicine Polyak K Lee MH Erdjument Bromage H Koff A Roberts JM Tempst P Massague J August 1994 Cloning of p27Kip1 a cyclin dependent kinase inhibitor and a potential mediator of extracellular antimitogenic signals Cell 78 1 59 66 doi 10 1016 0092 8674 94 90572 X PMID 8033212 S2CID 38513201 Chiarle R Pagano M Inghirami G 2001 The cyclin dependent kinase inhibitor p27 and its prognostic role in breast cancer Breast Cancer Research 3 2 91 4 doi 10 1186 bcr277 PMC 139437 PMID 11250752 Cooper Hausman Geoffrey Robert 2009 The Cell Boston University ASM Press p 669 ISBN 978 0 87893 300 6 Fero ML Rivkin M Tasch M Porter P Carow CE Firpo E Polyak K Tsai LH Broudy V Perlmutter RM Kaushansky K Roberts JM May 1996 A syndrome of multiorgan hyperplasia with features of gigantism tumorigenesis and female sterility in p27 Kip1 deficient mice Cell 85 5 733 44 doi 10 1016 S0092 8674 00 81239 8 PMID 8646781 S2CID 15490866 Kiyokawa H Kineman RD Manova Todorova KO Soares VC Hoffman ES Ono M Khanam D Hayday AC Frohman LA Koff A May 1996 Enhanced growth of mice lacking the cyclin dependent kinase inhibitor function of p27 Kip1 Cell 85 5 721 32 doi 10 1016 S0092 8674 00 81238 6 PMID 8646780 S2CID 19030818 Nakayama K Ishida N Shirane M Inomata A Inoue T Shishido N Horii I Loh DY Nakayama K May 1996 Mice lacking p27 Kip1 display increased body size multiple organ hyperplasia retinal dysplasia and pituitary tumors Cell 85 5 707 20 doi 10 1016 S0092 8674 00 81237 4 PMID 8646779 S2CID 2009281 Klopfleisch R Gruber AD January 2009 Differential expression of cell cycle regulators p21 p27 and p53 in metastasizing canine mammary adenocarcinomas versus normal mammary glands Res Vet Sci 87 1 91 6 doi 10 1016 j rvsc 2008 12 010 PMID 19185891 Klopfleisch R Schutze M Gruber AD September 2010 Loss of p27 expression in canine mammary tumors and their metastases Res Vet Sci 88 2 300 3 doi 10 1016 j rvsc 2009 08 007 PMID 19748645 Klopfleisch R von Euler H Sarli G Pinho SS Gartner F Gruber AD 2010 Molecular Carcinogenesis of Canine Mammary Tumors News From an Old Disease Veterinary Pathology 48 1 98 116 doi 10 1177 0300985810390826 PMID 21149845 S2CID 206509356 Klopfleisch R Schutze M Gruber AD October 2009 Downregulation of transforming growth factor b TGFb and latent TGFb binding protein LTBP 4 expression in late stage canine mammary tumours Veterinary Journal 186 3 379 84 doi 10 1016 j tvjl 2009 09 014 PMID 19836277 Gopfert U Kullmann M Hengst L July 2003 Cell cycle dependent translation of p27 involves a responsive element in its 5 UTR that overlaps with a uORF Hum Mol Genet 12 14 1767 79 doi 10 1093 hmg ddg177 PMID 12837699 a b c d e f g le Sage C Nagel R Agami R November 2007 Diverse ways to control p27Kip1 function miRNAs come into play Cell Cycle 6 22 2742 9 doi 10 4161 cc 6 22 4900 PMID 17986865 a b c d e f g h i j k l m n o p q r s t u v Chu IM Hengst L Slingerland JM April 2008 The Cdk inhibitor p27 in human cancer prognostic potential and relevance to anticancer therapy Nat Rev Cancer 8 4 253 67 doi 10 1038 nrc2347 PMID 18354415 S2CID 2175257 a b c d Nakayama KI Nakayama K May 2006 Ubiquitin ligases cell cycle control and cancer Nat Rev Cancer 6 5 369 81 doi 10 1038 nrc1881 PMID 16633365 S2CID 19594293 Kotoshiba S Kamura T Hara T Ishida N Nakayama KI May 2005 Molecular dissection of the interaction between p27 and Kip1 ubiquitylation promoting complex the ubiquitin ligase that regulates proteolysis of p27 in G1 phase J Biol Chem 280 18 17694 700 doi 10 1074 jbc M500866200 PMID 15746103 a b Ishida N Hara T Kamura T Yoshida M Nakayama K Nakayama KI April 2002 Phosphorylation of p27Kip1 on serine 10 is required for its binding to CRM1 and nuclear export J Biol Chem 277 17 14355 8 doi 10 1074 jbc C100762200 PMID 11889117 a b c d Chu I Sun J Arnaout A Kahn H Hanna W Narod S Sun P Tan CK Hengst L Slingerland J January 2007 p27 phosphorylation by Src regulates inhibition of cyclin E Cdk2 Cell 128 2 281 94 doi 10 1016 j cell 2006 11 049 PMC 1961623 PMID 17254967 McAllister SS Becker Hapak M Pintucci G Pagano M Dowdy SF January 2003 Novel p27 kip1 C terminal scatter domain mediates Rac dependent cell migration independent of cell cycle arrest functions Mol Cell Biol 23 1 216 28 doi 10 1128 MCB 23 1 216 228 2003 PMC 140659 PMID 12482975 Wu FY Wang SE Sanders ME Shin I Rojo F Baselga J Arteaga CL February 2006 Reduction of cytosolic p27 Kip1 inhibits cancer cell motility survival and tumorigenicity Cancer Res 66 4 2162 72 doi 10 1158 0008 5472 CAN 05 3304 PMID 16489017 Denicourt C Saenz CC Datnow B Cui XS Dowdy SF October 2007 Relocalized p27Kip1 tumor suppressor functions as a cytoplasmic metastatic oncogene in melanoma Cancer Res 67 19 9238 43 doi 10 1158 0008 5472 CAN 07 1375 PMID 17909030 Cheng M Sexl V Sherr CJ Roussel MF February 1998 Assembly of cyclin D dependent kinase and titration of p27Kip1 regulated by mitogen activated protein kinase kinase MEK1 Proc Natl Acad Sci U S A 95 3 1091 6 Bibcode 1998PNAS 95 1091C doi 10 1073 pnas 95 3 1091 PMC 18683 PMID 9448290 Liu X Sun Y Ehrlich M Lu T Kloog Y Weinberg RA Lodish HF Henis YI November 2000 Disruption of TGF beta growth inhibition by oncogenic ras is linked to p27Kip1 mislocalization Oncogene 19 51 5926 35 doi 10 1038 sj onc 1203991 PMID 11127824 Viglietto G Motti ML Bruni P Melillo RM D Alessio A Califano D Vinci F Chiappetta G Tsichlis P Bellacosa A Fusco A Santoro M October 2002 Cytoplasmic relocalization and inhibition of the cyclin dependent kinase inhibitor p27 Kip1 by PKB Akt mediated phosphorylation in breast cancer Nat Med 8 10 1136 44 doi 10 1038 nm762 PMID 12244303 S2CID 6580033 Larrea MD Hong F Wander SA da Silva TG Helfman D Lannigan D Smith JA Slingerland JM June 2009 RSK1 drives p27Kip1 phosphorylation at T198 to promote RhoA inhibition and increase cell motility Proc Natl Acad Sci U S A 106 23 9268 73 Bibcode 2009PNAS 106 9268L doi 10 1073 pnas 0805057106 PMC 2695095 PMID 19470470 a b Larrea MD Wander SA Slingerland JM November 2009 p27 as Jekyll and Hyde regulation of cell cycle and cell motility Cell Cycle 8 21 3455 61 doi 10 4161 cc 8 21 9789 PMID 19829074 a b Baldassarre G Belletti B Nicoloso MS Schiappacassi M Vecchione A Spessotto P Morrione A Canzonieri V Colombatti A January 2005 p27 Kip1 stathmin interaction influences sarcoma cell migration and invasion Cancer Cell 7 1 51 63 doi 10 1016 j ccr 2004 11 025 PMID 15652749 Chappuis PO Kapusta L Begin LR Wong N Brunet JS Narod SA Slingerland J Foulkes WD December 2000 Germline BRCA1 2 mutations and p27 Kip1 protein levels independently predict outcome after breast cancer J Clin Oncol 18 24 4045 52 doi 10 1200 jco 2000 18 24 4045 PMID 11118465 Chang BL Zheng SL Isaacs SD Wiley KE Turner A Li G Walsh PC Meyers DA Isaacs WB Xu J March 2004 A polymorphism in the CDKN1B gene is associated with increased risk of hereditary prostate cancer Cancer Res 64 6 1997 9 doi 10 1158 0008 5472 CAN 03 2340 PMID 15026335 Alrezk R Hannah Shmouni F Stratakis CA August 2017 MEN4 and CDKN1B mutations the latest of the MEN syndromes Endocrine Related Cancer 24 10 T195 208 doi 10 1530 ERC 17 0243 PMC 5623937 PMID 28824003 Masciullo V Sgambato A Pacilio C Pucci B Ferrandina G Palazzo J Carbone A Cittadini A Mancuso S Scambia G Giordano A August 1999 Frequent loss of expression of the cyclin dependent kinase inhibitor p27 in epithelial ovarian cancer Cancer Res 59 15 3790 4 PMID 10446997 Catzavelos C Bhattacharya N Ung YC Wilson JA Roncari L Sandhu C Shaw P Yeger H Morava Protzner I Kapusta L Franssen E Pritchard KI Slingerland JM February 1997 Decreased levels of the cell cycle inhibitor p27Kip1 protein prognostic implications in primary breast cancer Nat Med 3 2 227 30 doi 10 1038 nm0297 227 PMID 9018244 S2CID 25460889 a b Loda M Cukor B Tam SW Lavin P Fiorentino M Draetta GF Jessup JM Pagano M February 1997 Increased proteasome dependent degradation of the cyclin dependent kinase inhibitor p27 in aggressive colorectal carcinomas Nat Med 3 2 231 4 doi 10 1038 nm0297 231 PMID 9018245 S2CID 3164478 a b Esposito V Baldi A De Luca A Groger AM Loda M Giordano GG Caputi M Baldi F Pagano M Giordano A August 1997 Prognostic role of the cyclin dependent kinase inhibitor p27 in non small cell lung cancer Cancer Res 57 16 3381 5 PMID 9270000 Tsihlias J Kapusta LR DeBoer G Morava Protzner I Zbieranowski I Bhattacharya N Catzavelos GC Klotz LH Slingerland JM February 1998 Loss of cyclin dependent kinase inhibitor p27Kip1 is a novel prognostic factor in localized human prostate adenocarcinoma Cancer Res 58 3 542 8 PMID 9458103 Oshita F Kameda Y Nishio K Tanaka G Yamada K Nomura I Nakayama H Noda K 2000 Increased expression levels of cyclin dependent kinase inhibitor p27 correlate with good responses to platinum based chemotherapy in non small cell lung cancer Oncol Rep 7 3 491 5 doi 10 3892 or 7 3 491 PMID 10767357 Porter PL Barlow WE Yeh IT Lin MG Yuan XP Donato E Sledge GW Shapiro CL Ingle JN Haskell CM Albain KS Roberts JM Livingston RB Hayes DF December 2006 p27 Kip1 and cyclin E expression and breast cancer survival after treatment with adjuvant chemotherapy J Natl Cancer Inst 98 23 1723 31 doi 10 1093 jnci djj467 PMC 2727647 PMID 17148774 a b Blain SW Scher HI Cordon Cardo C Koff A February 2003 p27 as a target for cancer therapeutics Cancer Cell 3 2 111 5 doi 10 1016 S1535 6108 03 00026 6 PMID 12620406 Lowenheim H Furness DN Kil J Zinn C Gultig K Fero ML Frost D Gummer AW Roberts JM Rubel EW Hackney CM Zenner HP 1999 Gene disruption of p27 Kip1 allows cell proliferation in the postnatal and adult organ of corti Proc Natl Acad Sci U S A 96 7 4084 8 Bibcode 1999PNAS 96 4084L doi 10 1073 pnas 96 7 4084 PMC 22424 PMID 10097167 Nakagawa T 2014 Strategies for developing novel therapeutics for sensorineural hearing loss Front Pharmacol 5 206 doi 10 3389 fphar 2014 00206 PMC 4165348 PMID 25278894 a b Fujita N Sato S Katayama K Tsuruo T 2002 Akt dependent phosphorylation of p27Kip1 promotes binding to 14 3 3 and cytoplasmic localization J Biol Chem 277 32 28706 13 doi 10 1074 jbc M203668200 PMID 12042314 a b Wang W Ungermannova D Chen L Liu X 2003 A negatively charged amino acid in Skp2 is required for Skp2 Cks1 interaction and ubiquitination of p27Kip1 J Biol Chem 278 34 32390 6 doi 10 1074 jbc M305241200 PMID 12813041 a b Sitry D Seeliger MA Ko TK Ganoth D Breward SE Itzhaki LS Pagano M Hershko A 2002 Three different binding sites of Cks1 are required for p27 ubiquitin ligation J Biol Chem 277 44 42233 40 doi 10 1074 jbc M205254200 PMID 12140288 a b Lin J Jinno S Okayama H 2001 Cdk6 cyclin D3 complex evades inhibition by inhibitor proteins and uniquely controls cell s proliferation competence Oncogene 20 16 2000 9 doi 10 1038 sj onc 1204375 PMID 11360184 Rual JF Venkatesan K Hao T Hirozane Kishikawa T Dricot A Li N Berriz GF Gibbons FD Dreze M Ayivi Guedehoussou N Klitgord N Simon C Boxem M Milstein S Rosenberg J Goldberg DS Zhang LV Wong SL Franklin G Li S Albala JS Lim J Fraughton C Llamosas E Cevik S Bex C Lamesch P Sikorski RS Vandenhaute J Zoghbi HY Smolyar A Bosak S Sequerra R Doucette Stamm L Cusick ME Hill DE Roth FP Vidal M 2005 Towards a proteome scale map of the human protein protein interaction network Nature 437 7062 1173 8 Bibcode 2005Natur 437 1173R doi 10 1038 nature04209 PMID 16189514 S2CID 4427026 Zhang Q Wang X Wolgemuth DJ 1999 Developmentally regulated expression of cyclin D3 and its potential in vivo interacting proteins during murine gametogenesis Endocrinology 140 6 2790 800 doi 10 1210 endo 140 6 6756 PMID 10342870 a b c Connor MK Kotchetkov R Cariou S Resch A Lupetti R Beniston RG Melchior F Hengst L Slingerland JM 2003 CRM1 Ran mediated nuclear export of p27 Kip1 involves a nuclear export signal and links p27 export and proteolysis Mol Biol Cell 14 1 201 13 doi 10 1091 mbc E02 06 0319 PMC 140238 PMID 12529437 Shanahan F Seghezzi W Parry D Mahony D Lees E 1999 Cyclin E associates with BAF155 and BRG1 components of the mammalian SWI SNF complex and alters the ability of BRG1 to induce growth arrest Mol Cell Biol 19 2 1460 9 doi 10 1128 mcb 19 2 1460 PMC 116074 PMID 9891079 a b Porter LA Kong Beltran M Donoghue DJ 2003 Spy1 interacts with p27Kip1 to allow G1 S progression Mol Biol Cell 14 9 3664 74 doi 10 1091 mbc E02 12 0820 PMC 196558 PMID 12972555 Youn CK Cho HJ Kim SH Kim HB Kim MH Chang IY Lee JS Chung MH Hahm KS You HJ 2005 Bcl 2 expression suppresses mismatch repair activity through inhibition of E2F transcriptional activity Nat Cell Biol 7 2 137 47 doi 10 1038 ncb1215 PMID 15619620 S2CID 42766325 Law BK Chytil A Dumont N Hamilton EG Waltner Law ME Aakre ME Covington C Moses HL 2002 Rapamycin potentiates transforming growth factor beta induced growth arrest in nontransformed oncogene transformed and human cancer cells Mol Cell Biol 22 23 8184 98 doi 10 1128 MCB 22 23 8184 8198 2002 PMC 134072 PMID 12417722 Rosner M Hengstschlager M 2004 Tuberin binds p27 and negatively regulates its interaction with the SCF component Skp2 J Biol Chem 279 47 48707 15 doi 10 1074 jbc M405528200 PMID 15355997 Cariou S Donovan JC Flanagan WM Milic A Bhattacharya N Slingerland JM 2000 Down regulation of p21WAF1 CIP1 or p27Kip1 abrogates antiestrogen mediated cell cycle arrest in human breast cancer cells Proc Natl Acad Sci U S A 97 16 9042 6 Bibcode 2000PNAS 97 9042C doi 10 1073 pnas 160016897 PMC 16818 PMID 10908655 Sugiyama Y Tomoda K Tanaka T Arata Y Yoneda Kato N Kato J 2001 Direct binding of the signal transducing adaptor Grb2 facilitates down regulation of the cyclin dependent kinase inhibitor p27Kip1 J Biol Chem 276 15 12084 90 doi 10 1074 jbc M010811200 PMID 11278754 Smitherman M Lee K Swanger J Kapur R Clurman BE 2000 Characterization and targeted disruption of murine Nup50 a p27 Kip1 interacting component of the nuclear pore complex Mol Cell Biol 20 15 5631 42 doi 10 1128 MCB 20 15 5631 5642 2000 PMC 86029 PMID 10891500 Ishida N Hara T Kamura T Yoshida M Nakayama K Nakayama KI 2002 Phosphorylation of p27Kip1 on serine 10 is required for its binding to CRM1 and nuclear export J Biol Chem 277 17 14355 8 doi 10 1074 jbc C100762200 PMID 11889117 Further reading editMarone M Bonanno G Rutella S Leone G Scambia G Pierelli L 2003 Survival and cell cycle control in early hematopoiesis role of bcl 2 and the cyclin dependent kinase inhibitors P27 and P21 Leuk Lymphoma 43 1 51 7 doi 10 1080 10428190210195 PMID 11908736 S2CID 28490341 Hirabayashi H 2003 P27 expression and survival in NSCLC Nippon Rinsho 60 Suppl 5 263 6 PMID 12101669 Bloom J Pagano M 2003 Deregulated degradation of the cdk inhibitor p27 and malignant transformation Semin Cancer Biol 13 1 41 7 CiteSeerX 10 1 1 513 177 doi 10 1016 S1044 579X 02 00098 6 PMID 12507555 Tokumoto M Tsuruya K Fukuda K Kanai H Kuroki S Hirakata H Iida M 2003 Parathyroid cell growth in patients with advanced secondary hyperparathyroidism vitamin D receptor and cyclin dependent kinase inhibitors p21 and p27 Nephrol Dial Transplant 18 Suppl 3 90003 iii9 12 doi 10 1093 ndt gfg1003 PMID 12771291 Drexler HC 2004 The role of p27Kip1 in proteasome inhibitor induced apoptosis Cell Cycle 2 5 438 41 doi 10 4161 cc 2 5 461 PMID 12963837 Le XF Pruefer F Bast RC 2006 HER2 targeting antibodies modulate the cyclin dependent kinase inhibitor p27Kip1 via multiple signaling pathways Cell Cycle 4 1 87 95 doi 10 4161 cc 4 1 1360 PMID 15611642 Belletti B Nicoloso MS Schiappacassi M Chimienti E Berton S Lovat F Colombatti A Baldassarre G 2005 p27 kip1 functional regulation in human cancer a potential target for therapeutic designs Curr Med Chem 12 14 1589 605 doi 10 2174 0929867054367149 PMID 16022660 Sankaranarayanan P Schomay TE Aiello KA Alter O April 2015 Tensor GSVD of Patient and Platform Matched Tumor and Normal DNA Copy Number Profiles Uncovers Chromosome Arm Wide Patterns of Tumor Exclusive Platform Consistent Alterations Encoding for Cell Transformation and Predicting Ovarian Cancer Survival PLOS ONE 10 4 e0121396 Bibcode 2015PLoSO 1021396S doi 10 1371 journal pone 0121396 PMC 4398562 PMID 25875127 External links editCDKN1B human gene location in the UCSC Genome Browser CDKN1B human gene details in the UCSC Genome Browser Overview of all the structural information available in the PDB for UniProt P46527 Cyclin dependent kinase inhibitor 1B at the PDBe KB Retrieved from https en wikipedia org w index php title CDKN1B amp oldid 1171080158, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.