Difference between revisions of "Phosphatase Subfamily DSP6"

From PhosphataseWiki
Jump to: navigation, search
Line 8: Line 8:
 
DSP6 subfamily has two domains: rhodanese domain and phosphatase domain.  
 
DSP6 subfamily has two domains: rhodanese domain and phosphatase domain.  
  
Rhodanese domain inhibit phosphatase domain activity in DUSP6 <cite>Camps98</cite>. stabilizes the inactive conformation of the catalytic site and implies that the N-terminal domain functions as an allosteric inhibitor of phosphatase activity.  
+
Rhodanese domain inhibit phosphatase domain activity in DUSP6 <cite>Camps98</cite>, which is achieved by the binding of rhodanese domain and phosphatase domain. The binding stabilizes the inactive conformation of the phosphatase catalytic site <cite>Mark08</cite>. The rhodanese domain also mediates interaction with MAP kinases (often ERK) ('''via kinase interaction motif?'''). Its binding to MAP kinases induces conformation change in phosphatase domain, which can increase the phosphatase activity <cite>Stewart99</cite>.
  
Rhodanese domain mediates interaction with MAP kinases (often ERK). Rhodanese domain of DSP6 also has two conserved Leu-rich nuclear export signals <cite>Karlsson04</cite> (particular Figure 5 and Figure 11). Binding to MAP kinases induces conformation change in phosphatase domain, which can increase the phosphatase activity <cite>Stewart99</cite>.
+
Rhodanese domain of DSP6 also has two conserved Leu-rich nuclear export signals <cite>Karlsson04</cite> (particular Figure 5 and Figure 11).  
  
 
=== Function ===
 
=== Function ===
Line 17: Line 17:
 
DUSP6 preferentially dephosphorylates ERK <cite>Muda96, Groom96, Kim03</cite>, which resulted from that DUSP6 binds to ERK but not p38 or JNK. The interaction is mediated by rhodanese domain (or kinase interaction motif embedded in rhodanese domain?) <cite>Muda98</cite>. Later study has shown DUSP6 is ERK1/2-specific, as it does not inactive ERK5 <cite>Arkell08</cite>.
 
DUSP6 preferentially dephosphorylates ERK <cite>Muda96, Groom96, Kim03</cite>, which resulted from that DUSP6 binds to ERK but not p38 or JNK. The interaction is mediated by rhodanese domain (or kinase interaction motif embedded in rhodanese domain?) <cite>Muda98</cite>. Later study has shown DUSP6 is ERK1/2-specific, as it does not inactive ERK5 <cite>Arkell08</cite>.
  
As a negative regulator of ERK, DUSP6 is proposed to be tumor suppressor. DUSP6 expression was down-regulated through hypermethylation at enhancer in some pancreatic cell lines and pancreatic cancer tissues <cite>Furukawa98, Xu05</cite>. But, DUSP6 was up-regulated in endometrial adenocarcinomas <cite>Zhang13</cite>, thyroid carcinoma <cite>Lee12, DeglInnocenti13</cite>, and glioblastomas <cite>Messina11</cite>. Meanwhile, DUSP6 upregulation induced by angiotensin II mediates endothelial cell apoptosis <cite>Rossig02</cite>. (note: DUSP6 upregulation in cancer may be the response to suppress carcinogenesis?)
+
Furthermore, DUSP6, ERK2, and phosphorylated p38alpha can form a stable ternary complex in solution, and the phosphatase activity of DUSP6 toward p38alpha substrate is allosterically regulated by ERK2-DUSP6 interaction. This suggests that DUSP6 may mediate cross-talk between ERK and p38 pathways <cite>Zhang11</cite>.
 +
 
 +
As a negative regulator of ERK <cite>Muda96, Camps98, Jurek09, Zhang10</cite>, DUSP6 is proposed to be tumor suppressor via feedback mechanisms <cite>Zeliadt08</cite>:
 +
* DUSP6 plays tumor suppressive role in non-small-cell lung cancers <cite>Zhang10</cite>.
 +
* DUSP6 expression was correlated with lower histological grade and lower [http://en.wikipedia.org/wiki/Ki-67_%28protein%29 Ki-67 index] in the lung adenocarcinomas <cite>Lee11</cite>.
 +
* DUSP6 expression was down-regulated through hypermethylation at enhancer in some pancreatic cell lines and pancreatic cancer tissues <cite>Furukawa98, Xu05</cite>.  
 +
* Degradation of DUSP6 caused by reactive oxygen species (ROS) leads to aberrant ERK1/2 activation and contributes to tumorigenicity and chemoresistance of human ovarian cancer cells <cite>Chan08</cite>.
 +
* DUSP6 upregulation induced by angiotensin II mediates endothelial cell apoptosis <cite>Rossig02</cite>.
 +
 
 +
But, DUSP6 is up-regulated or not associated with cancers in some cases:
 +
* DUSP6 was up-regulated in endometrial adenocarcinomas <cite>Zhang13</cite>, thyroid carcinoma <cite>Lee12, DeglInnocenti13</cite>, and glioblastomas <cite>Messina11</cite>, MCF-7 breast cancer cells <cite>Nunes-Xavier10</cite>.
 +
* DUSP6 methylation is a rare event in endometrial cancer. Thus, silencing of the DUSP6 phosphatase is unlikely to contribute to constitutive activation of the ERK kinase cascade in endometrial cancer <cite>Chiappinelli10</cite>.
 +
* DUSP6 expression was not correlated with [http://en.wikipedia.org/wiki/Ki-67_%28protein%29 Ki-67 index] lung squamous cell carcinomas <cite>Lee11</cite>.
 +
 
 +
('''note: DUSP6 upregulation in cancer could be explained by feedback mechanisms?''')
  
 
Like DUSP2, DUSP4 and DUSP4 of DSP1 subfamily, DUSP6 is phosphorylated by ERK <cite>Marchetti05</cite>. However, the phosphorylation sites are different. DUSP6 is phosphorylated at serines 159 and 197 <cite>Marchetti05</cite>, which are found in DUSP6 and DUSP7, but not DUSP9 or other DSPs.
 
Like DUSP2, DUSP4 and DUSP4 of DSP1 subfamily, DUSP6 is phosphorylated by ERK <cite>Marchetti05</cite>. However, the phosphorylation sites are different. DUSP6 is phosphorylated at serines 159 and 197 <cite>Marchetti05</cite>, which are found in DUSP6 and DUSP7, but not DUSP9 or other DSPs.
  
Nitric oxide down-regulates MKP-3 mRNA levels <cite>Rossig00</cite>.
+
DUSP6 expression is regulated by:
 +
* [ETS-1].
 +
* [http://en.wikipedia.org/wiki/WT1 Wilms tumor protein (WT1)], a transcription factor as tumor suppressor, up-regulates DUSP6 expression <cite>Morrison08</cite>.
 +
* Nitric oxide down-regulates DUSP6 mRNA levels <cite>Rossig00</cite>.
  
 
====== DUSP7 (MKPX/PYST2) ======
 
====== DUSP7 (MKPX/PYST2) ======
Line 37: Line 54:
 
#Arkell08 pmid=18280112
 
#Arkell08 pmid=18280112
 
#Camps98 pmid=9596579
 
#Camps98 pmid=9596579
 +
#Chan08 pmid=18632752
 +
#Chiappinelli10 pmid=20638106
 
#DeglInnocenti13 pmid=23132790
 
#DeglInnocenti13 pmid=23132790
 
#Dickinson11 pmid=21908610
 
#Dickinson11 pmid=21908610
Line 42: Line 61:
 
#Furukawa98 pmid=9858808
 
#Furukawa98 pmid=9858808
 
#Groom96 pmid=8670865
 
#Groom96 pmid=8670865
 +
#Jurek09 pmid=19106095
 
#Karlsson04 pmid=15269220
 
#Karlsson04 pmid=15269220
 
#Kim03 pmid=14690430
 
#Kim03 pmid=14690430
 +
#Lee11 pmid=21680106
 
#Lee12 pmid=22535643
 
#Lee12 pmid=22535643
 
#Levy-Nissenbaum03a pmid=14576828
 
#Levy-Nissenbaum03a pmid=14576828
Line 50: Line 71:
 
#Liu07 pmid=18006813
 
#Liu07 pmid=18006813
 
#Marchetti05 pmid=15632084
 
#Marchetti05 pmid=15632084
 +
#Mark08 pmid=18694935
 
#Messina11 pmid=21499306
 
#Messina11 pmid=21499306
 +
#Morrison08 pmid=18644985
 
#Muda96 pmid=8626780
 
#Muda96 pmid=8626780
 
#Muda97 pmid=9030581
 
#Muda97 pmid=9030581
 
#Muda98 pmid=9535927
 
#Muda98 pmid=9535927
 +
#Nunes-Xavier10 pmid=20554528
 
#Rossig00 pmid=10846176
 
#Rossig00 pmid=10846176
 
#Rossig02 pmid=11998972
 
#Rossig02 pmid=11998972
Line 59: Line 83:
 
#Wu11 pmid=21943117
 
#Wu11 pmid=21943117
 
#Xu05 pmid=15824892
 
#Xu05 pmid=15824892
 +
#Zeliadt08 pmid=18771677
 +
#Zhang10 pmid=20097731
 +
#Zhang11 pmid=21454500
 
#Zhang13 pmid=23419500
 
#Zhang13 pmid=23419500
 
</biblio>
 
</biblio>

Revision as of 19:29, 7 March 2015

Phosphatase Classification: Fold CC1: Superfamily CC1: Family DSP: Subfamily DSP6

Evolution

Domain

DSP6 subfamily has two domains: rhodanese domain and phosphatase domain.

Rhodanese domain inhibit phosphatase domain activity in DUSP6 [1], which is achieved by the binding of rhodanese domain and phosphatase domain. The binding stabilizes the inactive conformation of the phosphatase catalytic site [2]. The rhodanese domain also mediates interaction with MAP kinases (often ERK) (via kinase interaction motif?). Its binding to MAP kinases induces conformation change in phosphatase domain, which can increase the phosphatase activity [3].

Rhodanese domain of DSP6 also has two conserved Leu-rich nuclear export signals [4] (particular Figure 5 and Figure 11).

Function

DUSP6 (MKP3/PYST1)

DUSP6 preferentially dephosphorylates ERK [5, 6, 7], which resulted from that DUSP6 binds to ERK but not p38 or JNK. The interaction is mediated by rhodanese domain (or kinase interaction motif embedded in rhodanese domain?) [8]. Later study has shown DUSP6 is ERK1/2-specific, as it does not inactive ERK5 [9].

Furthermore, DUSP6, ERK2, and phosphorylated p38alpha can form a stable ternary complex in solution, and the phosphatase activity of DUSP6 toward p38alpha substrate is allosterically regulated by ERK2-DUSP6 interaction. This suggests that DUSP6 may mediate cross-talk between ERK and p38 pathways [10].

As a negative regulator of ERK [1, 5, 11, 12], DUSP6 is proposed to be tumor suppressor via feedback mechanisms [13]:

  • DUSP6 plays tumor suppressive role in non-small-cell lung cancers [12].
  • DUSP6 expression was correlated with lower histological grade and lower Ki-67 index in the lung adenocarcinomas [14].
  • DUSP6 expression was down-regulated through hypermethylation at enhancer in some pancreatic cell lines and pancreatic cancer tissues [15, 16].
  • Degradation of DUSP6 caused by reactive oxygen species (ROS) leads to aberrant ERK1/2 activation and contributes to tumorigenicity and chemoresistance of human ovarian cancer cells [17].
  • DUSP6 upregulation induced by angiotensin II mediates endothelial cell apoptosis [18].

But, DUSP6 is up-regulated or not associated with cancers in some cases:

  • DUSP6 was up-regulated in endometrial adenocarcinomas [19], thyroid carcinoma [20, 21], and glioblastomas [22], MCF-7 breast cancer cells [23].
  • DUSP6 methylation is a rare event in endometrial cancer. Thus, silencing of the DUSP6 phosphatase is unlikely to contribute to constitutive activation of the ERK kinase cascade in endometrial cancer [24].
  • DUSP6 expression was not correlated with Ki-67 index lung squamous cell carcinomas [14].

(note: DUSP6 upregulation in cancer could be explained by feedback mechanisms?)

Like DUSP2, DUSP4 and DUSP4 of DSP1 subfamily, DUSP6 is phosphorylated by ERK [25]. However, the phosphorylation sites are different. DUSP6 is phosphorylated at serines 159 and 197 [25], which are found in DUSP6 and DUSP7, but not DUSP9 or other DSPs.

DUSP6 expression is regulated by:

  • [ETS-1].
  • Wilms tumor protein (WT1), a transcription factor as tumor suppressor, up-regulates DUSP6 expression [26].
  • Nitric oxide down-regulates DUSP6 mRNA levels [27].
DUSP7 (MKPX/PYST2)

DUSP7 is constitutively expressed in a wide variety of human cell lines. DUSP7 is predominantly cytosolic when expressed in COS-1 cells. In common with other members of DSP6 subfamily, DUSP7 shows substrate selectivity ERK > p38 = JNK. DUSP7 binds ERK in vivo. Both ERK and JNK activate DUSP7 phosphatase activity in vitro [28].

DUSP7 has at least two isoforms. The longer isoform is constitutively highly expressed in myeloid leukemia and other malignant cells [29, 30, 31].

DUSP9 (MKP4)

DUSP6 blocks activation of MAP kinases with the selectivity ERK > p38 = JNK. Same as other members in the subfamily, it locates in cytosol [32, 33]. DUSP9 is unique among these cytoplasmic MKPs in containing a conserved PKA consensus phosphorylation site (55)RRXSer-58 immediately adjacent to the kinase interaction motif. DUSP9 is phosphorylated on Ser-58 by PKA in vitro, and phosphorylation abrogates the binding of DUSP9 to both ERK2 and p38alpha MAP kinases [34].

Decreased expression of DUSP-9 is associated with poor prognosis in clear cell renal cell carcinomas [35].

References

  1. Camps M, Nichols A, Gillieron C, Antonsson B, Muda M, Chabert C, Boschert U, and Arkinstall S. Catalytic activation of the phosphatase MKP-3 by ERK2 mitogen-activated protein kinase. Science. 1998 May 22;280(5367):1262-5. DOI:10.1126/science.280.5367.1262 | PubMed ID:9596579 | HubMed [Camps98]
  2. Mark JK, Aubin RA, Smith S, and Hefford MA. Inhibition of mitogen-activated protein kinase phosphatase 3 activity by interdomain binding. J Biol Chem. 2008 Oct 17;283(42):28574-83. DOI:10.1074/jbc.M801747200 | PubMed ID:18694935 | HubMed [Mark08]
  3. Stewart AE, Dowd S, Keyse SM, and McDonald NQ. Crystal structure of the MAPK phosphatase Pyst1 catalytic domain and implications for regulated activation. Nat Struct Biol. 1999 Feb;6(2):174-81. DOI:10.1038/5861 | PubMed ID:10048930 | HubMed [Stewart99]
  4. Karlsson M, Mathers J, Dickinson RJ, Mandl M, and Keyse SM. Both nuclear-cytoplasmic shuttling of the dual specificity phosphatase MKP-3 and its ability to anchor MAP kinase in the cytoplasm are mediated by a conserved nuclear export signal. J Biol Chem. 2004 Oct 1;279(40):41882-91. DOI:10.1074/jbc.M406720200 | PubMed ID:15269220 | HubMed [Karlsson04]
  5. Muda M, Boschert U, Dickinson R, Martinou JC, Martinou I, Camps M, Schlegel W, and Arkinstall S. MKP-3, a novel cytosolic protein-tyrosine phosphatase that exemplifies a new class of mitogen-activated protein kinase phosphatase. J Biol Chem. 1996 Feb 23;271(8):4319-26. DOI:10.1074/jbc.271.8.4319 | PubMed ID:8626780 | HubMed [Muda96]
  6. Groom LA, Sneddon AA, Alessi DR, Dowd S, and Keyse SM. Differential regulation of the MAP, SAP and RK/p38 kinases by Pyst1, a novel cytosolic dual-specificity phosphatase. EMBO J. 1996 Jul 15;15(14):3621-32. PubMed ID:8670865 | HubMed [Groom96]
  7. Kim Y, Rice AE, and Denu JM. Intramolecular dephosphorylation of ERK by MKP3. Biochemistry. 2003 Dec 30;42(51):15197-207. DOI:10.1021/bi035346b | PubMed ID:14690430 | HubMed [Kim03]
  8. Muda M, Theodosiou A, Gillieron C, Smith A, Chabert C, Camps M, Boschert U, Rodrigues N, Davies K, Ashworth A, and Arkinstall S. The mitogen-activated protein kinase phosphatase-3 N-terminal noncatalytic region is responsible for tight substrate binding and enzymatic specificity. J Biol Chem. 1998 Apr 10;273(15):9323-9. DOI:10.1074/jbc.273.15.9323 | PubMed ID:9535927 | HubMed [Muda98]
  9. Arkell RS, Dickinson RJ, Squires M, Hayat S, Keyse SM, and Cook SJ. DUSP6/MKP-3 inactivates ERK1/2 but fails to bind and inactivate ERK5. Cell Signal. 2008 May;20(5):836-43. DOI:10.1016/j.cellsig.2007.12.014 | PubMed ID:18280112 | HubMed [Arkell08]
  10. Zhang YY, Wu JW, and Wang ZX. Mitogen-activated protein kinase (MAPK) phosphatase 3-mediated cross-talk between MAPKs ERK2 and p38alpha. J Biol Chem. 2011 May 6;286(18):16150-62. DOI:10.1074/jbc.M110.203786 | PubMed ID:21454500 | HubMed [Zhang11]
  11. Jurek A, Amagasaki K, Gembarska A, Heldin CH, and Lennartsson J. Negative and positive regulation of MAPK phosphatase 3 controls platelet-derived growth factor-induced Erk activation. J Biol Chem. 2009 Feb 13;284(7):4626-34. DOI:10.1074/jbc.M808490200 | PubMed ID:19106095 | HubMed [Jurek09]
  12. Zhang Z, Kobayashi S, Borczuk AC, Leidner RS, Laframboise T, Levine AD, and Halmos B. Dual specificity phosphatase 6 (DUSP6) is an ETS-regulated negative feedback mediator of oncogenic ERK signaling in lung cancer cells. Carcinogenesis. 2010 Apr;31(4):577-86. DOI:10.1093/carcin/bgq020 | PubMed ID:20097731 | HubMed [Zhang10]
  13. Zeliadt NA, Mauro LJ, and Wattenberg EV. Reciprocal regulation of extracellular signal regulated kinase 1/2 and mitogen activated protein kinase phosphatase-3. Toxicol Appl Pharmacol. 2008 Nov 1;232(3):408-17. DOI:10.1016/j.taap.2008.08.007 | PubMed ID:18771677 | HubMed [Zeliadt08]
  14. Lee H, Kim JM, Huang SM, Park SK, Kim DH, Kim DH, Lee CS, Suh KS, Yi ES, and Kim KH. Differential expression of DUSP6 with expression of ERK and Ki-67 in non-small cell lung carcinoma. Pathol Res Pract. 2011 Jul 15;207(7):428-32. DOI:10.1016/j.prp.2011.05.004 | PubMed ID:21680106 | HubMed [Lee11]
  15. Furukawa T, Yatsuoka T, Youssef EM, Abe T, Yokoyama T, Fukushige S, Soeda E, Hoshi M, Hayashi Y, Sunamura M, Kobari M, and Horii A. Genomic analysis of DUSP6, a dual specificity MAP kinase phosphatase, in pancreatic cancer. Cytogenet Cell Genet. 1998;82(3-4):156-9. DOI:10.1159/000015091 | PubMed ID:9858808 | HubMed [Furukawa98]
  16. Xu S, Furukawa T, Kanai N, Sunamura M, and Horii A. Abrogation of DUSP6 by hypermethylation in human pancreatic cancer. J Hum Genet. 2005;50(4):159-167. DOI:10.1007/s10038-005-0235-y | PubMed ID:15824892 | HubMed [Xu05]
  17. Chan DW, Liu VW, Tsao GS, Yao KM, Furukawa T, Chan KK, and Ngan HY. Loss of MKP3 mediated by oxidative stress enhances tumorigenicity and chemoresistance of ovarian cancer cells. Carcinogenesis. 2008 Sep;29(9):1742-50. DOI:10.1093/carcin/bgn167 | PubMed ID:18632752 | HubMed [Chan08]
  18. Rössig L, Hermann C, Haendeler J, Assmus B, Zeiher AM, and Dimmeler S. Angiotensin II-induced upregulation of MAP kinase phosphatase-3 mRNA levels mediates endothelial cell apoptosis. Basic Res Cardiol. 2002 Jan;97(1):1-8. DOI:10.1007/s395-002-8381-2 | PubMed ID:11998972 | HubMed [Rossig02]
  19. Zhang H, Guo Q, Wang C, Yan L, Fu Y, Fan M, Zhao X, and Li M. Dual-specificity phosphatase 6 (Dusp6), a negative regulator of FGF2/ERK1/2 signaling, enhances 17β-estradiol-induced cell growth in endometrial adenocarcinoma cell. Mol Cell Endocrinol. 2013 Aug 25;376(1-2):60-9. DOI:10.1016/j.mce.2013.02.007 | PubMed ID:23419500 | HubMed [Zhang13]
  20. Lee JU, Huang S, Lee MH, Lee SE, Ryu MJ, Kim SJ, Kim YK, Kim SY, Joung KH, Kim JM, Shong M, and Jo YS. Dual specificity phosphatase 6 as a predictor of invasiveness in papillary thyroid cancer. Eur J Endocrinol. 2012 Jul;167(1):93-101. DOI:10.1530/EJE-12-0010 | PubMed ID:22535643 | HubMed [Lee12]
  21. Degl'Innocenti D, Romeo P, Tarantino E, Sensi M, Cassinelli G, Catalano V, Lanzi C, Perrone F, Pilotti S, Seregni E, Pierotti MA, Greco A, and Borrello MG. DUSP6/MKP3 is overexpressed in papillary and poorly differentiated thyroid carcinoma and contributes to neoplastic properties of thyroid cancer cells. Endocr Relat Cancer. 2013 Feb;20(1):23-37. DOI:10.1530/ERC-12-0078 | PubMed ID:23132790 | HubMed [DeglInnocenti13]
  22. Messina S, Frati L, Leonetti C, Zuchegna C, Di Zazzo E, Calogero A, and Porcellini A. Dual-specificity phosphatase DUSP6 has tumor-promoting properties in human glioblastomas. Oncogene. 2011 Sep 1;30(35):3813-20. DOI:10.1038/onc.2011.99 | PubMed ID:21499306 | HubMed [Messina11]
  23. Nunes-Xavier CE, Tárrega C, Cejudo-Marín R, Frijhoff J, Sandin A, Ostman A, and Pulido R. Differential up-regulation of MAP kinase phosphatases MKP3/DUSP6 and DUSP5 by Ets2 and c-Jun converge in the control of the growth arrest versus proliferation response of MCF-7 breast cancer cells to phorbol ester. J Biol Chem. 2010 Aug 20;285(34):26417-30. DOI:10.1074/jbc.M110.121830 | PubMed ID:20554528 | HubMed [Nunes-Xavier10]
  24. Chiappinelli KB, Rimel BJ, Massad LS, and Goodfellow PJ. Infrequent methylation of the DUSP6 phosphatase in endometrial cancer. Gynecol Oncol. 2010 Oct;119(1):146-50. DOI:10.1016/j.ygyno.2010.06.015 | PubMed ID:20638106 | HubMed [Chiappinelli10]
  25. Marchetti S, Gimond C, Chambard JC, Touboul T, Roux D, Pouysségur J, and Pagès G. Extracellular signal-regulated kinases phosphorylate mitogen-activated protein kinase phosphatase 3/DUSP6 at serines 159 and 197, two sites critical for its proteasomal degradation. Mol Cell Biol. 2005 Jan;25(2):854-64. DOI:10.1128/MCB.25.2.854-864.2005 | PubMed ID:15632084 | HubMed [Marchetti05]
  26. Morrison DJ, Kim MK, Berkofsky-Fessler W, and Licht JD. WT1 induction of mitogen-activated protein kinase phosphatase 3 represents a novel mechanism of growth suppression. Mol Cancer Res. 2008 Jul;6(7):1225-31. DOI:10.1158/1541-7786.MCR-08-0078 | PubMed ID:18644985 | HubMed [Morrison08]
  27. Rössig L, Haendeler J, Hermann C, Malchow P, Urbich C, Zeiher AM, and Dimmeler S. Nitric oxide down-regulates MKP-3 mRNA levels: involvement in endothelial cell protection from apoptosis. J Biol Chem. 2000 Aug 18;275(33):25502-7. DOI:10.1074/jbc.M002283200 | PubMed ID:10846176 | HubMed [Rossig00]
  28. Dowd S, Sneddon AA, and Keyse SM. Isolation of the human genes encoding the pyst1 and Pyst2 phosphatases: characterisation of Pyst2 as a cytosolic dual-specificity MAP kinase phosphatase and its catalytic activation by both MAP and SAP kinases. J Cell Sci. 1998 Nov;111 ( Pt 22):3389-99. DOI:10.1242/jcs.111.22.3389 | PubMed ID:9788880 | HubMed [Dowd98]
  29. Levy-Nissenbaum O, Sagi-Assif O, Kapon D, Hantisteanu S, Burg T, Raanani P, Avigdor A, Ben-Bassat I, and Witz IP. Dual-specificity phosphatase Pyst2-L is constitutively highly expressed in myeloid leukemia and other malignant cells. Oncogene. 2003 Oct 23;22(48):7649-60. DOI:10.1038/sj.onc.1206971 | PubMed ID:14576828 | HubMed [Levy-Nissenbaum03a]
  30. Levy-Nissenbaum O, Sagi-Assif O, Raanani P, Avigdor A, Ben-Bassat I, and Witz IP. cDNA microarray analysis reveals an overexpression of the dual-specificity MAPK phosphatase PYST2 in acute leukemia. Methods Enzymol. 2003;366:103-13. DOI:10.1016/s0076-6879(03)66009-x | PubMed ID:14674243 | HubMed [Levy-Nissenbaum03b]
  31. Levy-Nissenbaum O, Sagi-Assif O, and Witz IP. Characterization of the dual-specificity phosphatase PYST2 and its transcripts. Genes Chromosomes Cancer. 2004 Jan;39(1):37-47. DOI:10.1002/gcc.10295 | PubMed ID:14603440 | HubMed [Levy-Nissenbaum04]
  32. Muda M, Boschert U, Smith A, Antonsson B, Gillieron C, Chabert C, Camps M, Martinou I, Ashworth A, and Arkinstall S. Molecular cloning and functional characterization of a novel mitogen-activated protein kinase phosphatase, MKP-4. J Biol Chem. 1997 Feb 21;272(8):5141-51. DOI:10.1074/jbc.272.8.5141 | PubMed ID:9030581 | HubMed [Muda97]
  33. Liu Y, Lagowski J, Sundholm A, Sundberg A, and Kulesz-Martin M. Microtubule disruption and tumor suppression by mitogen-activated protein kinase phosphatase 4. Cancer Res. 2007 Nov 15;67(22):10711-9. DOI:10.1158/0008-5472.CAN-07-1968 | PubMed ID:18006813 | HubMed [Liu07]
  34. Dickinson RJ, Delavaine L, Cejudo-Marín R, Stewart G, Staples CJ, Didmon MP, Trinidad AG, Alonso A, Pulido R, and Keyse SM. Phosphorylation of the kinase interaction motif in mitogen-activated protein (MAP) kinase phosphatase-4 mediates cross-talk between protein kinase A and MAP kinase signaling pathways. J Biol Chem. 2011 Nov 4;286(44):38018-38026. DOI:10.1074/jbc.M111.255844 | PubMed ID:21908610 | HubMed [Dickinson11]
  35. Wu S, Wang Y, Sun L, Zhang Z, Jiang Z, Qin Z, Han H, Liu Z, Li X, Tang A, Gui Y, Cai Z, and Zhou F. Decreased expression of dual-specificity phosphatase 9 is associated with poor prognosis in clear cell renal cell carcinoma. BMC Cancer. 2011 Sep 26;11:413. DOI:10.1186/1471-2407-11-413 | PubMed ID:21943117 | HubMed [Wu11]
All Medline abstracts: PubMed | HubMed