109 results on '"Nick Morrice"'
Search Results
2. Identification of the amino acids 300-600 of IRS-2 as 14-3-3 binding region with the importance of IGF-1/insulin-regulated phosphorylation of Ser-573.
- Author
-
Sabine S Neukamm, Rachel Toth, Nick Morrice, David G Campbell, Carol Mackintosh, Rainer Lehmann, Hans-Ulrich Haering, Erwin D Schleicher, and Cora Weigert
- Subjects
Medicine ,Science - Abstract
Phosphorylation of insulin receptor substrate (IRS)-2 on tyrosine residues is a key event in IGF-1/insulin signaling and leads to activation of the PI 3-kinase and the Ras/MAPK pathway. Furthermore, phosphorylated serine/threonine residues on IRS-2 can induce 14-3-3 binding. In this study we searched IRS-2 for novel phosphorylation sites and investigated the interaction between IRS-2 and 14-3-3. Mass spectrometry identified a total of 24 serine/threonine residues on IRS-2 with 12 sites unique for IRS-2 while the other residues are conserved in IRS-1 and IRS-2. IGF-1 stimulation led to increased binding of 14-3-3 to IRS-2 in transfected HEK293 cells and this binding was prevented by inhibition of the PI 3-kinase pathway and an Akt/PKB inhibitor. Insulin-stimulated interaction between endogenous IRS-2 and 14-3-3 was observed in rat hepatoma cells and in mice liver after an acute insulin stimulus and refeeding. Using different IRS-2 fragments enabled localization of the IGF-1-dependent 14-3-3 binding region spanning amino acids 300-600. The 24 identified residues on IRS-2 included several 14-3-3 binding candidates in the region 300-600. Single alanine mutants of these candidates led to the identification of serine 573 as 14-3-3 binding site. A phospho-site specific antibody was generated to further characterize serine 573. IGF-1-dependent phosphorylation of serine 573 was reduced by inhibition of PI 3-kinase and Akt/PKB. A negative role of this phosphorylation site was implicated by the alanine mutant of serine 573 which led to enhanced phosphorylation of Akt/PKB in an IGF-1 time course experiment. To conclude, our data suggest a physiologically relevant role for IGF-1/insulin-dependent 14-3-3 binding to IRS-2 involving serine 573.
- Published
- 2012
- Full Text
- View/download PDF
3. Isolation of human mitotic protein phosphatase complexes: identification of a complex between protein phosphatase 1 and the RNA helicase Ddx21.
- Author
-
Veerle De Wever, David C Lloyd, Isha Nasa, Mhairi Nimick, Laura Trinkle-Mulcahy, Robert Gourlay, Nick Morrice, and Greg B G Moorhead
- Subjects
Medicine ,Science - Abstract
Metazoan mitosis requires remodelling of sub-cellular structures to ensure proper division of cellular and genetic material. Faults often lead to genomic instability, cell cycle arrests and disease onset. These key structural changes are under tight spatial-temporal and post-translational control, with crucial roles for reversible protein phosphorylation. The phosphoprotein phosphatases PP1 and PP2A are paramount for the timely execution of mitotic entry and exit but their interaction partners and substrates are still largely unresolved. High throughput, mass-spectrometry based studies have limited sensitivity for the detection of low-abundance and transient complexes, a typical feature of many protein phosphatase complexes. Moreover, the limited timeframe during which mitosis takes place reduces the likelihood of identifying mitotic phosphatase complexes in asynchronous cells. Hence, numerous mitotic protein phosphatase complexes still await identification. Here we present a strategy to enrich and identify serine/threonine protein phosphatase complexes at the mitotic spindle. We thus identified a nucleolar RNA helicase, Ddx21/Gu, as a novel, direct PP1 interactor. Furthermore, our results place PP1 within the toposome, a Topoisomerase II alpha (TOPOIIα) containing complex with a key role in mitotic chromatin regulation and cell cycle progression, possibly via regulated protein phosphorylation. This study provides a strategy for the identification of further mitotic PP1 partners and the unravelling of PP1 functions during mitosis.
- Published
- 2012
- Full Text
- View/download PDF
4. Regulation of Human PINK1 ubiquitin kinase by Serine167, Serine228 and Cysteine412 phosphorylation
- Author
-
Andrew D. Waddell, Hina Ojha, Shalini Agarwal, Christopher J. Clarke, Ana Terriente-Felix, Houjiang Zhou, Poonam Kakade, Axel Knebel, Andrew M. Shaw, Robert Gourlay, Joby Varghese, Renata F. Soares, Rachel Toth, Thomas Macartney, Patrick A. Eyers, Nick Morrice, Richard Bayliss, Alexander J. Whitworth, Claire E. Eyers, and Miratul M. K. Muqit
- Abstract
Loss-of-function mutations in the human PINK1 kinase (hPINK1) are causative of early-onset Parkinson’s disease (PD). Activation of hPINK1 induces phosphorylated ubiquitin to initiate removal of damaged mitochondria by autophagy. Previously we solved the structure of the insect PINK1 orthologue, Tribolium castaneum PINK1, and showed that autophosphorylation of Ser205 was critical for ubiquitin interaction and phosphorylation (Kumar, Tamjar, Waddell et al., 2017). In this advance, we report new findings on the regulation of hPINK1 by phosphorylation. We reconstitute E. coli expressed hPINK1 activity in vitro by direct incorporation of phosphoserine at the equivalent site Serine 228 (pSer228), providing direct evidence for a role for Ser228 phosphorylation in hPINK1 activation. Furthermore, using mass spectrometry, we identify six novel Ser/Thr autophosphorylation sites including regulatory Serine167 phosphorylation (pSer167), which in addition to pSer228 is required for ubiquitin recognition and phosphorylation. Strikingly, we also detect phosphorylation of a conserved Cysteine412 (pCys412) residue in the hPINK1 activation segment. Structural modelling suggests that pCys412 inhibits ubiquitin recognition and we demonstrate that mutation of Cys412 to Ala renders hPINK1 more active towards ubiquitin when expressed in human cells. These results outline new insights into hPINK1 activation by pSer167 and pSer228 and a novel inhibitory mechanism mediated by pCys412. These findings will aid in the development of small molecule activators of hPINK1. Description of files: Figure 1 Figure 1c OPA1 Coomassie: First gel: +Phos Tag Lane 1-2: WT, Lane 3-4 pS228 recombinant PINK1 Second gel: -Phos Tag Lane 1-2: WT, Lane 3-4 pS228 recombinant PINK1 Figure 1c blot: Lane 1-2: WT, Lane 3-4 pS228 recombinant PINK1 Figure 1d Coomassie: Recombinant PINK1: Lane 1-2: WT, Lane 3-4: KI, Lane 5-6 pS228, Lane 7-8: pS228DIns3, Lane 9-10: pS228G309D Figure 1d autorad: Recombinant PINK1: Lane 1-2: WT, Lane 3-4: KI, Lane 5-6 pS228, Lane 7-8: pS228DIns3, Lane 9-10: pS228G309D Figure 1f FLAG: Lane 1: Marker, Lane 2: empty, Lane 3-4: FLAG-empty (-CCCP), Lane 5-6: FLAG-empty (+CCCP), Lane 7-8: hPINK1 WT (-CCCP), Lane 9-10: hPINK1 WT (+CCCP), Lane 11-12: hPINK1 KI (-CCCP), Lane 13-14: hPINK1 KI (+CCCP), Lane 15-16: hPINK1 S228A (-CCCP), Lane 17-18: hPINK1 S228A (+CCCP) Figure 1f HSP60: Lane 1: Marker, Lane 2: empty, Lane 3-4: FLAG-empty (-CCCP), Lane 5-6: FLAG-empty (+CCCP), Lane 7-8: hPINK1 WT (-CCCP), Lane 9-10: hPINK1 WT (+CCCP), Lane 11-12: hPINK1 KI (-CCCP), Lane 13-14: hPINK1 KI (+CCCP), Lane 15-16: hPINK1 S228A (-CCCP), Lane 17-18: hPINK1 S228A (+CCCP) Figure 1f OPA1: Lane 1: Marker, Lane 2: empty, Lane 3-4: FLAG-empty (-CCCP), Lane 5-6: FLAG-empty (+CCCP), Lane 7-8: hPINK1 WT (-CCCP), Lane 9-10: hPINK1 WT (+CCCP), Lane 11-12: hPINK1 KI (-CCCP), Lane 13-14: hPINK1 KI (+CCCP), Lane 15-16: hPINK1 S228A (-CCCP), Lane 17-18: hPINK1 S228A (+CCCP) Figure 1f pS228: Lane 1: Marker, Lane 2: empty, Lane 3-4: FLAG-empty (-CCCP), Lane 5-6: FLAG-empty (+CCCP), Lane 7-8: hPINK1 WT (-CCCP), Lane 9-10: hPINK1 WT (+CCCP), Lane 11-12: hPINK1 KI (-CCCP), Lane 13-14: hPINK1 KI (+CCCP), Lane 15-16: hPINK1 S228A (-CCCP), Lane 17-18: hPINK1 S228A (+CCCP) Figure 1f PINK1 pS228 FLAG merge: Lane 1: Marker, Lane 2: empty, Lane 3-4: FLAG-empty (-CCCP), Lane 5-6: FLAG-empty (+CCCP), Lane 7-8: hPINK1 WT (-CCCP), Lane 9-10: hPINK1 WT (+CCCP), Lane 11-12: hPINK1 KI (-CCCP), Lane 13-14: hPINK1 KI (+CCCP), Lane 15-16: hPINK1 S228A (-CCCP), Lane 17-18: hPINK1 S228A (+CCCP) Figure 1g PINK1 pS228 PINK1 multiplex: Lane 1: Marker, Lane 2-3: WT SK-OV-3 (-O/A), Lane 4-5: WT SK-OV-3 (+O/A), Lane 6-7: KO PINK1 SK-OV-3 (-O/A), Lane 8-9: KO PINK1 SK-OV-3 (+O/A) Figure 1g pS228: Lane 1: Marker, Lane 2-3: WT SK-OV-3 (-O/A), Lane 4-5: WT SK-OV-3 (+O/A), Lane 6-7: KO PINK1 SK-OV-3 (-O/A), Lane 8-9: KO PINK1 SK-OV-3 (+O/A) Figure 1g PINK1: Lane 1: Marker, Lane 2-3: WT SK-OV-3 (-O/A), Lane 4-5: WT SK-OV-3 (+O/A), Lane 6-7: KO PINK1 SK-OV-3 (-O/A), Lane 8-9: KO PINK1 SK-OV-3 (+O/A) Figure 1g Tom20: Lane 1: Marker, Lane 2-3: WT SK-OV-3 (-O/A), Lane 4-5: WT SK-OV-3 (+O/A), Lane 6-7: KO PINK1 SK-OV-3 (-O/A), Lane 8-9: KO PINK1 SK-OV-3 (+O/A) Figure 1h HSP60: Upper blot Lane 1: Marker, Lane 2-3: emp FLAG (-CCCP), Lane 4-5: emp FLAG (+CCCP), Lane 6-7: WT PINK1-FLAG (-CCCP), Lane 8-9: WT PINK1-FLAG (+CCCP), Lane10-11: KI PINK1-FLAG (-CCCP), Lane 12-13: KI PINK1-FLAG (+CCCP), Lane14-15: S228A PINK1-FLAG (-CCCP), Lane 16-17: S228A PINK1-FLAG (+CCCP) Lower blot Lane 1: Marker, Lane 2-3: S228N PINK1 FLAG (-CCCP), Lane 4-5: S228N PINK1 FLAG (+CCCP), Lane 6-7: S230A PINK1-FLAG (-CCCP), Lane 8-9: S230A PINK1-FLAG (+CCCP), Lane10-11: DIns3 PINK1-FLAG (-CCCP), Lane 12-13: DIns3 PINK1-FLAG (+CCCP), Rest of the lanes: Extra samples not included in the paper. Figure 1h OPA1: Upper blot Lane 1: Marker, Lane 2-3: emp FLAG (-CCCP), Lane 4-5: emp FLAG (+CCCP), Lane 6-7: WT PINK1-FLAG (-CCCP), Lane 8-9: WT PINK1-FLAG (+CCCP), Lane10-11: KI PINK1-FLAG (-CCCP), Lane 12-13: KI PINK1-FLAG (+CCCP), Lane14-15: S228A PINK1-FLAG (-CCCP), Lane 16-17: S228A PINK1-FLAG (+CCCP) Lower blot Lane 1: Marker, Lane 2-3: S228N PINK1 FLAG (-CCCP), Lane 4-5: S228N PINK1 FLAG (+CCCP), Lane 6-7: S230A PINK1-FLAG (-CCCP), Lane 8-9: S230A PINK1-FLAG (+CCCP), Lane10-11: DIns3 PINK1-FLAG (-CCCP), Lane 12-13: DIns3 PINK1-FLAG (+CCCP) Figure 1h pS228 PINK1: Upper blot Lane 1: Marker, Lane 2-3: emp FLAG (-CCCP), Lane 4-5: emp FLAG (+CCCP), Lane 6-7: WT PINK1-FLAG (-CCCP), Lane 8-9: WT PINK1-FLAG (+CCCP), Lane10-11: KI PINK1-FLAG (-CCCP), Lane 12-13: KI PINK1-FLAG (+CCCP), Lane14-15: S228A PINK1-FLAG (-CCCP), Lane 16-17: S228A PINK1-FLAG (+CCCP) Lower blot Lane 1: Marker, Lane 2-3: S228N PINK1 FLAG (-CCCP), Lane 4-5: S228N PINK1 FLAG (+CCCP), Lane 6-7: S230A PINK1-FLAG (-CCCP), Lane 8-9: S230A PINK1-FLAG (+CCCP), Lane10-11: DIns3 PINK1-FLAG (-CCCP), Lane 12-13: DIns3 PINK1-FLAG (+CCCP) Figure 1h pUb: Upper blot Lane 1: Marker, Lane 2-3: emp FLAG (-CCCP), Lane 4-5: emp FLAG (+CCCP), Lane 6-7: WT PINK1-FLAG (-CCCP), Lane 8-9: WT PINK1-FLAG (+CCCP), Lane10-11: KI PINK1-FLAG (-CCCP), Lane 12-13: KI PINK1-FLAG (+CCCP), Lane14-15: S228A PINK1-FLAG (-CCCP), Lane 16-17: S228A PINK1-FLAG (+CCCP) Lower blot Lane 1: Marker, Lane 2-3: S228N PINK1 FLAG (-CCCP), Lane 4-5: S228N PINK1 FLAG (+CCCP), Lane 6-7: S230A PINK1-FLAG (-CCCP), Lane 8-9: S230A PINK1-FLAG (+CCCP), Lane10-11: DIns3 PINK1-FLAG (-CCCP), Lane 12-13: DIns3 PINK1-FLAG (+CCCP) Figure 1- Figure Supplement 2b: Recombinant jason chin post-purification. 2 lanes loaded per construct, one for amylose purification and another for the subsequent 6His purification. Lane1 and Lane5 are included in the paper. Lane 1 - WT PINK1, amylose (included in paper), Lane 2 - WT PINK1, 6His, Lane 3 - KI PINK1, amylose, Lane 4 - KI PINK1, 6His, Lane 5 - pSer228 PINK1, amylose (included in paper), Lane 6 - pSer228 PINK1, 6His, Lane 7 - pSer284 PINK1, amylose, Lane 8 - pSer284 PINK1, 6His, Lane 9 - pSer402 PINK1, amylose, Lane 10 - pSer402 PINK1, 6His Figure 1- Figure Supplement 2c: Lane10 and Lane11 are included in the paper. Lane 1 - 2mg/ml BSA, Lane 2 - 1mg/ml BSA, Lane 3 - 0.5mg/ml BSA, Lane 4 - 0.25mg/ml BSA, Lane 5 - 0.125 mg/ml BSA, Lane 6 – ladder, Lane 7 - unrelated PINK1 construct (Ser228A), Lane 8 - unrelated PINK1 construct (Ser284A), Lane 9 - KI PINK1, Lane 10 - WT PINK1, Lane 11 - pSer228 PINK1, 6His, Lane 12 - pSer284 PINK1, 6His Figure 1- Figure Supplement 2d (+ phos tag): Lane 1-2: WT recombinant PINK1, Lane 3-4: KI recombinant PINK1, Lane 5-6: pS228 recombinant PINK1, Lane 7-8: pSer284 recombinant PINK1 (Not included in the paper) Figure 1- Figure Supplement 2d (no phos tag): Lane 1-2: WT recombinant PINK1, Lane 3-4: KI recombinant PINK1, Lane 5-6: pS228 recombinant PINK1, Lane 7-8: pSer284 recombinant PINK1 (Not included in the paper) Figure 1- Figure Supplement 2f Coomassie: Recombinant PINK1: Lane 1-2: WT, Lane 3-4: KI, Lane 5-6 pS228, Lane 7-8: pS228DIns3, Lane 9-10: pS228G309D Figure 1 - Figure Supplement 2f autorad: Recombinant PINK1: Lane 1-2: WT, Lane 3-4: KI, Lane 5-6 pS228, Lane 7-8: pS228DIns3, Lane 9-10: pS228G309D Figure 1 - Figure Supplement 3: Immunofluorescence images as mentioned in the paper Figure 2 Figure 2a FLAG: Lane 1: Marker, Lane3-4: WT PINK1 3FLAG (-CCCP), Lane 5-6: WT PINK1 3FLAG (+CCCP), Lane 7-8: KI PINK1 3FLAG (-CCCP), Lane 9-10: KI PINK1 3FLAG (+CCCP) Figure 2a HSP60: Lane 1: Marker, Lane3-4: WT PINK1 3FLAG (-CCCP), Lane 5-6: WT PINK1 3FLAG (+CCCP), Lane 7-8: KI PINK1 3FLAG (-CCCP), Lane 9-10: KI PINK1 3FLAG (+CCCP), rest of the lanes: Extra samples not included in the paper Figure 2a OPA1: Lane 1: Marker, Lane3-4: WT PINK1 3FLAG (-CCCP), Lane 5-6: WT PINK1 3FLAG (+CCCP), Lane 7-8: KI PINK1 3FLAG (-CCCP), Lane 9-10: KI PINK1 3FLAG (+CCCP) Figure 2a Phostag: Lane 1: Marker, Lane3-4: WT PINK1 3FLAG (-CCCP), Lane 5-6: WT PINK1 3FLAG (+CCCP), Lane 7-8: KI PINK1 3FLAG (-CCCP), Lane 9-10: KI PINK1 3FLAG (+CCCP) Figure 2- Figure Supplement 1a: Lane 1: Marker, Lane 3: emp 3-FLAG (-CCCP), Lane 4: emp 3-FLAG (+CCCP), Lane 5: KI PINK1 3-FLAG (-CCCP), Lane 6: KI PINK1 3-FLAG (+CCCP), Lane 7: WT PINK1 3-FLAG (-CCCP), Lane 8: WT PINK1 3-FLAG (+CCCP), Rest of the lanes: Not included in the paper Figure 2- Figure Supplement 1b: Lane 1: Marker, Lane3-5: 1% TX-100 (Total, soluble, insoluble), Lane 6-8: 1% SDS (Total, soluble, insoluble), Lane 10-12: 2% SDS (Total, soluble, insoluble) Figure 2- Figure Supplement 1c: Lane 1: Marker, Lane3-5: WT DMSO (Input, SN, IP), Lane 6-8: WT CCCP (Input, SN, IP), Lane 10-12: KI DMSO (Input, SN, IP), Lane 14-16: KI CCCP (Input, SN, IP) Figure 2- Figure Supplement 1d DCP: Upper blot Lane 1: Marker, Lane3-4: SK-OV-3 PINK1 KO SDS (+CCCP), Lane 5-6: PINK1 KO Triton (+CCCP), Lane 7-8: PINK1 KO SDS of leftover pellet (+CCCP) Lower blot Lane 1: Marker, Lane3-4: SK-OV-3 WT SDS (-CCCP), Lane 5-6: SK-OV-3 WT Triton (-CCCP), Lane 7-8: SK-OV-3 WT SDS of leftover pellet (-CCCP), Lane9-10: SK-OV-3 WT SDS (+CCCP), Lane 11-12: SK-OV-3 WT Triton (+CCCP), Lane 13-14: SK-OV-3 WT SDS of leftover pellet (+CCCP) Figure 2- Figure Supplement 1d ps228: Upper blot Lane 1: Marker, Lane3-4: SK-OV-3 PINK1 KO SDS (+CCCP), Lane 5-6: PINK1 KO Triton (+CCCP), Lane 7-8: PINK1 KO SDS of leftover pellet (+CCCP) Lower blot Lane 1: Marker, Lane3-4: SK-OV-3 WT SDS (-CCCP), Lane 5-6: SK-OV-3 WT Triton (-CCCP), Lane 7-8: SK-OV-3 WT SDS of leftover pellet (-CCCP), Lane9-10: SK-OV-3 WT SDS (+CCCP), Lane 11-12: SK-OV-3 WT Triton (+CCCP), Lane 13-14: SK-OV-3 WT SDS of leftover pellet (+CCCP) Figure 2- Figure Supplement 1d mix: Upper blot Lane 1: Marker, Lane3-4: SK-OV-3 PINK1 KO SDS (+CCCP), Lane 5-6: PINK1 KO Triton (+CCCP), Lane 7-8: PINK1 KO SDS of leftover pellet (+CCCP) Lower blot Lane 1: Marker, Lane3-4: SK-OV-3 WT SDS (-CCCP), Lane 5-6: SK-OV-3 WT Triton (-CCCP), Lane 7-8: SK-OV-3 WT SDS of leftover pellet (-CCCP), Lane9-10: SK-OV-3 WT SDS (+CCCP), Lane 11-12: SK-OV-3 WT Triton (+CCCP), Lane 13-14: SK-OV-3 WT SDS of leftover pellet (+CCCP) Figure 2- Figure Supplement 1d HSP60 KO: Lane 1: Marker, Lane3-4: SK-OV-3 PINK1 KO SDS (+CCCP), Lane 5-6: PINK1 KO Triton (+CCCP), Lane 7-8: PINK1 KO SDS of leftover pellet (+CCCP) Figure 2- Figure Supplement 1d HSP60 WT: Lane 1: Marker, Lane3-4: SK-OV-3 WT SDS (-CCCP), Lane 5-6: SK-OV-3 WT Triton (-CCCP), Lane 7-8: SK-OV-3 WT SDS of leftover pellet (-CCCP), Lane9-10: SK-OV-3 WT SDS (+CCCP), Lane 11-12: SK-OV-3 WT Triton (+CCCP), Lane 13-14: SK-OV-3 WT SDS of leftover pellet (+CCCP) Figure 2- Figure Supplement 1d ACSL1 HSP60 KO: HSP60 blot (Lower band) re-probed for ACSL1 (Upper band); Lane 1: Marker, Lane3-4: SK-OV-3 PINK1 KO SDS (+CCCP), Lane 5-6: PINK1 KO Triton (+CCCP), Lane 7-8: PINK1 KO SDS of leftover pellet (+CCCP) Figure 2- Figure Supplement 1d ACSL1 HSP60 WT: HSP60 blot (Lower band) re-probed for ACSL1 (Upper band); Lane 1: Marker, Lane3-4: SK-OV-3 WT SDS (-CCCP), Lane 5-6: SK-OV-3 WT Triton (-CCCP), Lane 7-8: SK-OV-3 WT SDS of leftover pellet (-CCCP), Lane9-10: SK-OV-3 WT SDS (+CCCP), Lane 11-12: SK-OV-3 WT Triton (+CCCP), Lane 13-14: SK-OV-3 WT SDS of leftover pellet (+CCCP) Figure 2- Figure Supplement 1e: Lane 1: Marker, Lane 3: WT Trypsin (-CCCP), Lane 5: WT chymotrypsin (-CCCP), Lane 7: WT Trypsin (+CCCP), Lane 9: WT chymotrypsin (+CCCP), Lane 11: KI Trypsin (-CCCP), Lane 13: KI chymotrypsin (-CCCP), Lane 15: KI Trypsin (+CCCP), Lane 17: KI chymotrypsin (+CCCP) Figure 3 Figure 3c OPA1 PINK1: Upper blot: OPA1 Lane 1: Marker, Lane 2: empty, Lane 3-4: hPINK1 WT (-O/A), Lane 5-6: hPINK1 WT (+O/A), Lane 7-8: hPINK1 KI (-O/A), Lane 9-10: hPINK1 KI (+O/A), Lane 11-12: hPINK1 S228A (-O/A), Lane 13-14: hPINK1 S228A (+O/A), Lane 15-16: hPINK1 S167A (-O/A), Lane 17-18: hPINK1 S167A (+O/A), Lane 19-20: hPINK1 S228A+S167A (-O/A), Lane 21-22: hPINK1 S228A+S167A (+O/A), Lane 23-24: hPINK1 multi-mutant (-O/A), Lane 25-26: hPINK1 multi-mutant (+O/A) Lower blot: PINK1 Lane 1: Marker, Lane 2: empty, Lane 3-4: hPINK1 WT (-O/A), Lane 5-6: hPINK1 WT (+O/A), Lane 7-8: hPINK1 KI (-O/A), Lane 9-10: hPINK1 KI (+O/A), Lane 11-12: hPINK1 S228A (-O/A), Lane 13-14: hPINK1 S228A (+O/A), Lane 15-16: hPINK1 S167A (-O/A), Lane 17-18: hPINK1 S167A (+O/A), Lane 19-20: hPINK1 S228A+S167A (-O/A), Lane 21-22: hPINK1 S228A+S167A (+O/A), Lane 23-24: hPINK1 multi-mutant (-O/A), Lane 25-26: hPINK1 multi-mutant (+O/A) Figure 3c ps228 PINK1: Upper blot: pS228 PINK1 Lane 1: Marker, Lane 2: empty, Lane 3-4: hPINK1 WT (-O/A), Lane 5-6: hPINK1 WT (+O/A), Lane 7-8: hPINK1 KI (-O/A), Lane 9-10: hPINK1 KI (+O/A), Lane 11-12: hPINK1 S228A (-O/A), Lane 13-14: hPINK1 S228A (+O/A), Lane 15-16: hPINK1 S167A (-O/A), Lane 17-18: hPINK1 S167A (+O/A), Lane 19-20: hPINK1 S228A+S167A (-O/A), Lane 21-22: hPINK1 S228A+S167A (+O/A), Lane 23-24: hPINK1 multi-mutant (-O/A), Lane 25-26: hPINK1 multi-mutant (+O/A) Lower blot: HSP60 Lane 1: Marker, Lane 2: empty, Lane 3-4: hPINK1 WT (-O/A), Lane 5-6: hPINK1 WT (+O/A), Lane 7-8: hPINK1 KI (-O/A), Lane 9-10: hPINK1 KI (+O/A), Lane 11-12: hPINK1 S228A (-O/A), Lane 13-14: hPINK1 S228A (+O/A), Lane 15-16: hPINK1 S167A (-O/A), Lane 17-18: hPINK1 S167A (+O/A), Lane 19-20: hPINK1 S228A+S167A (-O/A), Lane 21-22: hPINK1 S228A+S167A (+O/A), Lane 23-24: hPINK1 multi-mutant (-O/A), Lane 25-26: hPINK1 multi-mutant (+O/A) Figure 3c pUb: Blot: pS65 Ub Lane 1: Marker, Lane 2: empty, Lane 3-4: hPINK1 WT (-O/A), Lane 5-6: hPINK1 WT (+O/A), Lane 7-8: hPINK1 KI (-O/A), Lane 9-10: hPINK1 KI (+O/A), Lane 11-12: hPINK1 S228A (-O/A), Lane 13-14: hPINK1 S228A (+O/A), Lane 15-16: hPINK1 S167A (-O/A), Lane 17-18: hPINK1 S167A (+O/A), Lane 19-20: hPINK1 S228A+S167A (-O/A), Lane 21-22: hPINK1 S228A+S167A (+O/A), Lane 23-24: hPINK1 multi-mutant (-O/A), Lane 25-26: hPINK1 multi-mutant (+O/A) Figure 4 Figure 4e PINK1: Full blot (cut according to kDa before probing with antibody); Lane 1: Marker, Lane 3: emp 3FLAG (-CCCP), Lane4: emp 3FLAG (+CCCP for 180 min), Lane 5: KI PINK 3FLAG (-CCCP), Lane6: KI PINK 3FLAG (+CCCP for 180 min), Lane 7: WT PINK 3FLAG (-CCCP), Lane 8: WT PINK 3FLAG (+CCCP for 15 min), Lane 9: WT PINK 3FLAG (+CCCP for 30 min), Lane 10: WT PINK 3FLAG (+CCCP for 45 min), Lane 11: WT PINK 3FLAG (+CCCP for 60 min), Lane 12: WT PINK 3FLAG (+CCCP for 120 min), Lane 13: WT PINK 3FLAG (+CCCP for 180 min), Lane 14: C412A PINK 3FLAG (-CCCP), Lane 15: C412A PINK 3FLAG (+CCCP for 15 min), Lane 16: C412A PINK 3FLAG (+CCCP for 30 min), Lane 17: C412A PINK 3FLAG (+CCCP for 45 min), Lane 18: C412A PINK 3FLAG (+CCCP for 60 min), Lane 19: C412A PINK 3FLAG (+CCCP for 120 min), Lane 20: C412A PINK 3FLAG (+CCCP for 180 min) Figure 4e GAPDH OPA: Upper bands: OPA1, Lower bands: GAPDH; Lane 1: Marker, Lane 3: emp 3FLAG (-CCCP), Lane4: emp 3FLAG (+CCCP for 180 min), Lane 5: KI PINK 3FLAG (-CCCP), Lane6: KI PINK 3FLAG (+CCCP for 180 min), Lane 7: WT PINK 3FLAG (-CCCP), Lane 8: WT PINK 3FLAG (+CCCP for 15 min), Lane 9: WT PINK 3FLAG (+CCCP for 30 min), Lane 10: WT PINK 3FLAG (+CCCP for 45 min), Lane 11: WT PINK 3FLAG (+CCCP for 60 min), Lane 12: WT PINK 3FLAG (+CCCP for 120 min), Lane 13: WT PINK 3FLAG (+C
- Published
- 2023
5. Expression of Concern: Protein phosphatase 4 interacts with the Survival of Motor Neurons complex and enhances the temporal localisation of snRNPs
- Author
-
Graeme K. Carnegie, Judith E. Sleeman, Nick Morrice, C. James Hastie, Mark W. Peggie, Amanda Philp, Angus I. Lamond, and Patricia T. W. Cohen
- Subjects
Cell Biology - Published
- 2022
6. PKC isoforms activate LRRK1 kinase by phosphorylating conserved residues (Ser1064, Ser1074 and Thr1075) within the CORB GTPase domain
- Author
-
Asad U Malik, Athanasios Karapetsas, Raja S. Nirujogi, Deep Chatterjee, Toan K. Phung, Melanie Wightman, Robert Gourlay, Nick Morrice, Sebastian Mathea, Stefan Knapp, and Dario R Alessi
- Abstract
Leucine-rich-repeat-kinase 1 (LRRK1) and its homologue LRRK2 are multidomain kinases possessing a ROC-CORA-CORB containing GTPase domain and phosphorylate distinct Rab proteins. LRRK1 loss of function mutations cause the bone disorder osteosclerotic metaphyseal dysplasia, whereas LRRK2 missense mutations that enhance kinase activity cause Parkinson’s disease. Previous work suggested that LRRK1 but not LRRK2, is activated via a Protein Kinase C (PKC)-dependent mechanism. Here we demonstrate that phosphorylation and activation of LRRK1 in HEK293 cells is blocked by PKC inhibitors including LXS-196 (Darovasertib), a compound that has entered clinical trials. We show multiple PKC isoforms phosphorylate and activate recombinant LRRK1 in a manner reversed by phosphatase treatment. PKCα unexpectedly does not activate LRRK1 by phosphorylating the kinase domain, but instead phosphorylates a cluster of conserved residues (Ser1064, Ser1074 and Thr1075) located within a region of the CORB domain of the GTPase domain. These residues are positioned at the equivalent region of the LRRK2 DK helix reported to stabilize the kinase domain αC-helix in the active conformation. Thr1075 represents an optimal PKC site phosphorylation motif and its mutation to Ala, blocked PKC-mediated activation of LRRK1. A triple Glu mutation of Ser1064/Ser1074/Thr1075 to mimic phosphorylation, enhanced LRRK1 kinase activity ~3-fold. From analysis of available structures, we postulate that phosphorylation of Ser1064, Ser1074 and Thr1075 activates LRRK1 by promoting interaction and stabilization of the aC-helix on the kinase domain. This study provides new fundamental insights into the mechanism controlling LRRK1 activity and reveals a novel unexpected activation mechanism.
- Published
- 2022
7. Accelerated Protein Biomarker Discovery from FFPE Tissue Samples Using Single-Shot, Short Gradient Microflow SWATH MS
- Author
-
Chunhui Yuan, Shuang Liang, Tiannan Guo, Guan Ruan, Christie L. Hunter, Xue Cai, Xiaoyan Yu, Nick Morrice, Weigang Ge, Chen Chen, Tiansheng Zhu, Lirong Chen, Yi Zhu, Shaozheng Dai, Qiushi Zhang, Zhongzhi Luan, Rui Sun, and Ruedi Aebersold
- Subjects
Male ,Proteomics ,0301 basic medicine ,Swath ms ,Chromatography ,030102 biochemistry & molecular biology ,Protein biomarkers ,Formalin fixed paraffin embedded ,Chemistry ,Single shot ,General Chemistry ,Batch effect ,Biochemistry ,03 medical and health sciences ,030104 developmental biology ,Humans ,Biomarker discovery ,Peptides ,Biomarkers ,Software - Abstract
We reported and evaluated a microflow, single-shot, short gradient SWATH MS method intended to accelerate the discovery and verification of protein biomarkers in preclassified clinical specimens. The method uses a 15 min gradient microflow-LC peptide separation, an optimized SWATH MS window configuration, and OpenSWATH software for data analysis. We applied the method to a cohort containing 204 FFPE tissue samples from 58 prostate cancer patients and 10 benign prostatic hyperplasia patients. Altogether we identified 27,975 proteotypic peptides and 4037 SwissProt proteins from these 204 samples. Compared to a reference SWATH method with a 2 h gradient, we found 3800 proteins were quantified by the two methods on two different instruments with relatively high consistency (r = 0.77). The accelerated method consumed only 17% instrument time, while quantifying 80% of proteins compared to the 2 h gradient SWATH. Although the missing value rate increased by 20%, batch effects reduced by 21%. 75 deregulated proteins measured by the accelerated method were selected for further validation. A shortlist of 134 selected peptide precursors from the 75 proteins were analyzed using MRM-HR, and the results exhibited high quantitative consistency with the 15 min SWATH method (r = 0.89) in the same sample set. We further verified the applicability of these 75 proteins in separating benign and malignant tissues (AUC = 0.99) in an independent prostate cancer cohort (n = 154). Altogether, the results showed that the 15 min gradient microflow SWATH accelerated large-scale data acquisition by 6 times, reduced batch effect by 21%, introduced 20% more missing values, and exhibited comparable ability to separate disease groups.
- Published
- 2020
8. Cov-MS
- Author
-
Dan Lane, Sigrid Verhelst, Maarten Dhaenens, Amy C. Harms, Griet Debyser, Nicolas Drouin, Johannes P. C. Vissers, Lize Cuypers, Katleen Van Uytfanghe, Dieter Deforce, Stuart A. Oehrle, Catherine S. Lane, Jan Claereboudt, Péter Judák, Nathan Debunne, Sally Hannam, Lennart Martens, Pathmanaban Ramasamy, Robbin Bouwmeester, Andrea Bhangu-Uhlmann, N. Leigh Anderson, Laurence Van Oudenhove, Nick Morrice, Sven Degroeve, Laura Corveleyn, Marc Cherlet, Peter Van Eenoo, Morteza Razavi, Tim Van Den Bossche, Evelien Wynendaele, Ruben t’Kindt, Said El Ouadi, Emmie Dumont, Nikunj Tanna, Bart De Spiegeleer, Laura De Clerck, Katrien Lagrou, Surya Gupta, Tim Reyns, Thomas Hankemeier, Pankaj Gupta, Christophe P. Stove, Bart Van Puyvelde, Donald J. L. Jones, Florian C. Sigloch, Simon Daled, Sander Willems, Olivier Tytgat, Ralf Gabriels, Jean-Baptiste Vincendet, Laurie De Wilde, Geert A. Martens, Steve Silvester, K. Roels, Koen Sandra, Department of Bio-engineering Sciences, Faculty of Sciences and Bioengineering Sciences, Pathology/molecular and cellular medicine, and Diabetes Pathology & Therapy
- Subjects
Proteomics ,Coronavirus disease 2019 (COVID-19) ,Computer science ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Chemistry, Multidisciplinary ,Economic shortage ,Spreading ,Computational biology ,Rising population density ,infectious diseases ,Protein detection ,Article ,Mass Spectrometry ,reverse transcription polymerase chain reaction ,03 medical and health sciences ,Viral Proteins ,Medicine and Health Sciences ,Global mobility ,QD1-999 ,Diagnostics ,030304 developmental biology ,Community based ,0303 health sciences ,Science & Technology ,Pandemic ,Biochemistry, Genetics and Molecular Biology(all) ,SARS-CoV-2 ,030302 biochemistry & molecular biology ,COVID-19 ,Diagnostic test ,global mobility ,QUANTIFICATION ,3. Good health ,Chemistry ,Physical Sciences ,MRM - Abstract
Rising population density and global mobility are among the reasons why pathogens such as SARS-CoV-2, the virus that causes COVID-19, spread so rapidly across the globe. The policy response to such pandemics will always have to include accurate monitoring of the spread, as this provides one of the few alternatives to total lockdown. However, COVID-19 diagnosis is currently performed almost exclusively by reverse transcription polymerase chain reaction (RT-PCR). Although this is efficient, automatable, and acceptably cheap, reliance on one type of technology comes with serious caveats, as illustrated by recurring reagent and test shortages. We therefore developed an alternative diagnostic test that detects proteolytically digested SARS-CoV-2 proteins using mass spectrometry (MS). We established the Cov-MS consortium, consisting of 15 academic laboratories and several industrial partners to increase applicability, accessibility, sensitivity, and robustness of this kind of SARS-CoV-2 detection. This, in turn, gave rise to the Cov-MS Digital Incubator that allows other laboratories to join the effort, navigate, and share their optimizations and translate the assay into their clinic. As this test relies on viral proteins instead of RNA, it provides an orthogonal and complementary approach to RT-PCR using other reagents that are relatively inexpensive and widely available, as well as orthogonally skilled personnel and different instruments. Data are available via ProteomeXchange with identifier PXD022550. ispartof: JACS AU vol:1 issue:6 pages:750-765 ispartof: location:United States status: published
- Published
- 2021
9. Human protein phosphatase 5 dissociates from heat-shock proteins and is proteolytically activated in response to arachidonic acid and the microtubule-depolymerizing drug nocodazole
- Author
-
Nick Morrice, Patricia T.W. Cohen, Cristina Vázquez-Martin, and Tamás Zeke
- Subjects
Cytoplasm ,Phosphatase ,Antineoplastic Agents ,Biology ,Microtubules ,Models, Biological ,Biochemistry ,Cell Line ,chemistry.chemical_compound ,Heat shock protein ,Phosphoprotein Phosphatases ,Humans ,Nuclear protein ,Molecular Biology ,Heat-Shock Proteins ,Cell Nucleus ,Arachidonic Acid ,Nocodazole ,Cell Cycle ,Nuclear Proteins ,Protein phosphatase 2 ,Cell Biology ,Transport protein ,Cell biology ,Hsp70 ,Enzyme Activation ,Molecular Weight ,Protein Transport ,chemistry ,Proteasome ,Mutation ,Protein Processing, Post-Translational ,Protein Binding ,Research Article - Abstract
Ppp5 (protein phosphatase 5) is a serine/threonine protein phosphatase that has been conserved throughout eukaryotic evolution. In mammalian cells, FLAG-tagged Ppp5 and endogenous Ppp5 are found to interact with endogenous Hsp (heat-shock protein) 70, as well as Hsp90. Incubation of cells with arachidonic acid or the microtubule-depolymerizing agent, nocodazole, causes loss of interaction of Hsp70 and Hsp90 with FLAG-tagged Ppp5 and increase of Ppp5 activity. In response to the same treatments, endogenous Ppp5 undergoes proteolytic cleavage of the N- and C-termini, with the subsequent appearance of high-molecular-mass species. The results indicate that Ppp5 is activated by proteolysis on dissociation from Hsps, and is destroyed via the proteasome after ubiquitination. Cleavage at the C-terminus removes a nuclear localization sequence, allowing these active cleaved forms of Ppp5 to translocate to the cytoplasm. The response of Ppp5 to arachidonic acid and nocodazole suggests that Ppp5 may be required for stress-related processes that can sometimes cause cell-cycle arrest, and leads to the first description for in vivo regulation of Ppp5 activity.
- Published
- 2021
10. Accelerated Protein Biomarker Discovery from FFPE tissue samples using Single-shot, Short Gradient Microflow SWATH MS
- Author
-
Xue Cai, Xiaoyan Yu, Rui Sun, Weigang Ge, Chunhui Yuan, Shuang Liang, Chen Chen, Yi Zhu, Tiannan Guo, Shaozheng Dai, Christie L. Hunter, Lirong Chen, Zhongzhi Luan, Qiushi Zhang, Nick Morrice, and Ruedi Aebersold
- Subjects
chemistry.chemical_classification ,Swath ms ,Chromatography ,medicine.anatomical_structure ,Protein biomarkers ,Formalin fixed paraffin embedded ,Chemistry ,Prostate ,Single shot ,medicine ,Peptide ,Batch effect ,Biomarker discovery - Abstract
We report and evaluated a microflow, single-shot, short gradient SWATH MS method intended to accelerate the discovery and verification of protein biomarkers in clinical specimens. The method uses 15-min gradient microflow-LC peptide separation, an optimized SWATH MS window configuration and OpenSWATH software for data analysis.We applied the method to a cohort 204 of FFPE prostate tissue samples from 58 prostate cancer patients and 10 prostatic hyperplasia patients. Altogether we identified 27,976 proteotypic peptides and 4,043 SwissProt proteins from these 204 samples. Compared to a reference SWATH method with 2-hour gradient the accelerated method consumed only 27% instrument time, quantified 80% proteins and showed reduced batch effects. 3,800 proteins were quantified by both methods in two different instruments with relatively high consistency (r = 0.77). 75 proteins detected by the accelerated method with differential abundance between clinical groups were selected for further validation. A shortlist of 134 selected peptide precursors from the 75 proteins were analyzed using MRM-HR, exhibiting high quantitative consistency with the 15-min SWATH method (r = 0.89) in the same sample set. We further verified the capacity of these 75 proteins in separating benign and malignant tissues (AUC = 0.99) in an independent prostate cancer cohort (n=154).Overall our data show that the single-shot short gradient microflow-LC SWATH MS method achieved about 4-fold acceleration of data acquisition with reduced batch effect and a moderate level of protein attrition compared to a standard SWATH acquisition method. Finally, the results showed comparable ability to separate clinical groups.
- Published
- 2019
- Full Text
- View/download PDF
11. Accelerating SWATH® Acquisition for Protein Quantitation – Up to 100 Samples per Day
- Author
-
Nick Morrice, Randy J. Arnold, Christie L. Hunter, and Zuzana Demianova
- Subjects
Chromatography ,Chemistry ,Quantitative proteomics ,Genetics ,Molecular Biology ,Biochemistry ,Biotechnology - Published
- 2020
12. Identification of sites phosphorylated by the vaccinia virus B1R kinase in viral protein H5R
- Author
-
Brown, Neil G., Nick Morrice, D, Beaud, Georges, Hardie, Grahame, and Leader, David P
- Published
- 2000
- Full Text
- View/download PDF
13. Depletion of Myofibril-Associated Proteins Using Selective Protein Extraction as a Tool in Cardiac Proteomics
- Author
-
Achim, Treumann, Pawel, Palmowski, Wing Chiu, Tong, Julie, Taggart, Nick, Morrice, G, Nicholas Europe-Finner, and Michael J, Taggart
- Subjects
Proteomics ,Myocardium ,Guinea Pigs ,Microfilament Proteins ,Muscle Proteins ,Sodium Dodecyl Sulfate ,Buffers ,Myofibrils ,Tandem Mass Spectrometry ,Animals ,Trypsin ,Edetic Acid ,Software ,Chromatography, Liquid - Abstract
Muscle tissue poses a particular challenge to proteomic analysis due to a very wide range of protein abundances arising from the dominant expression of myofilament-related proteins. We address this issue by describing proteomic analysis with liquid chromatography-mass spectrometry (LC-MS) and sequential window acquisition of all theoretical mass spectra (SWATH), of guinea pig cardiac tissue prepared in two homogenization buffers: (1) An SDS-based buffer designed to extract "all" tissue proteins and (2) a long-established EDTA-containing buffer thought to preferentially extract non-myofibril-related proteins. We use gene ontology (GO) annotation-based assessment of subcellular localization to indicate if these enriched proteins congregate in the cytoplasm or in organellar lumens. This technique results in the preferential quantitation of less abundant non-myofibrillar proteins and, for future studies, offers the opportunity for more complete analyses of changes in heart tissue protein expression with biological circumstance.
- Published
- 2017
14. LPP3 mediates self-generation of chemotactic LPA gradients by melanoma cells
- Author
-
Sergey Tumanov, Matthew Nielson, Gillian M. Mackay, Peter A. Thomason, Luke Tweedy, Nick Morrice, Andrew J. Muinonen-Martin, Robert H. Insall, Olivia Susanto, Jurre J. Kamphorst, and Yvette W. H. Koh
- Subjects
0301 basic medicine ,Skin Neoplasms ,LPP3 ,Phosphatase ,Cell ,Phosphatidate Phosphatase ,Biology ,Metastasis ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Line, Tumor ,Lysophosphatidic acid ,medicine ,Humans ,Neoplasm Invasiveness ,Melanoma ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Gene knockdown ,Chemotaxis ,Cell Biology ,medicine.disease ,Cell biology ,LPA ,Self-generated gradients ,030104 developmental biology ,medicine.anatomical_structure ,Enzyme ,Biochemistry ,chemistry ,030220 oncology & carcinogenesis ,lipids (amino acids, peptides, and proteins) ,Autotaxin ,biological phenomena, cell phenomena, and immunity ,Lysophospholipids ,Research Article - Abstract
Melanoma cells steer out of tumours using self-generated lysophosphatidic acid (LPA) gradients. The cells break down LPA, which is present at high levels around the tumours, creating a dynamic gradient that is low in the tumour and high outside. They then migrate up this gradient, creating a complex and evolving outward chemotactic stimulus. Here, we introduce a new assay for self-generated chemotaxis, and show that raising LPA levels causes a delay in migration rather than loss of chemotactic efficiency. Knockdown of the lipid phosphatase LPP3 – but not of its homologues LPP1 or LPP2 – diminishes the cell's ability to break down LPA. This is specific for chemotactically active LPAs, such as the 18:1 and 20:4 species. Inhibition of autotaxin-mediated LPA production does not diminish outward chemotaxis, but loss of LPP3-mediated LPA breakdown blocks it. Similarly, in both 2D and 3D invasion assays, knockdown of LPP3 diminishes the ability of melanoma cells to invade. Our results demonstrate that LPP3 is the key enzyme in the breakdown of LPA by melanoma cells, and confirm the importance of attractant breakdown in LPA-mediated cell steering. This article has an associated First Person interview with the first author of the paper., Highlighted Article: Melanoma cells can create and follow their own gradients of attractant, via a new mechanism by which tumour cells may undergo metastasis.
- Published
- 2017
- Full Text
- View/download PDF
15. Depletion of Myofibril-Associated Proteins Using Selective Protein Extraction as a Tool in Cardiac Proteomics
- Author
-
Michael J. Taggart, G. Nicholas Europe-Finner, Pawel Palmowski, Julie Taggart, Achim Treumann, Nick Morrice, and Wing Chiu Tong
- Subjects
0301 basic medicine ,Muscle tissue ,Chemistry ,Cardiac muscle ,Proteomics ,Subcellular localization ,Molecular biology ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Cytoplasm ,Protein purification ,medicine ,Myofibril ,Homogenization (biology) - Abstract
Muscle tissue poses a particular challenge to proteomic analysis due to a very wide range of protein abundances arising from the dominant expression of myofilament-related proteins. We address this issue by describing proteomic analysis with liquid chromatography-mass spectrometry (LC-MS) and sequential window acquisition of all theoretical mass spectra (SWATH), of guinea pig cardiac tissue prepared in two homogenization buffers: (1) An SDS-based buffer designed to extract "all" tissue proteins and (2) a long-established EDTA-containing buffer thought to preferentially extract non-myofibril-related proteins. We use gene ontology (GO) annotation-based assessment of subcellular localization to indicate if these enriched proteins congregate in the cytoplasm or in organellar lumens. This technique results in the preferential quantitation of less abundant non-myofibrillar proteins and, for future studies, offers the opportunity for more complete analyses of changes in heart tissue protein expression with biological circumstance.
- Published
- 2017
16. IκB Kinase 2 Regulates TPL-2 Activation of Extracellular Signal-Regulated Kinases 1 and 2 by Direct Phosphorylation of TPL-2 Serine 400
- Author
-
Agnes Mambole-Dema, Huei Ting Yang, Steven C. Ley, Derek W. Abbott, Julia Janzen, Abduelhakem Ben-Addi, Nick Morrice, Thorsten Gantke, and Karine Roget
- Subjects
Lipopolysaccharides ,Transcription, Genetic ,Biology ,Mitogen-activated protein kinase kinase ,Transfection ,MAP2K7 ,Mice ,TANK-binding kinase 1 ,Proto-Oncogene Proteins ,Serine ,Animals ,Humans ,c-Raf ,Phosphorylation ,Molecular Biology ,MAPK14 ,Mice, Knockout ,Mitogen-Activated Protein Kinase 1 ,Serine/threonine-specific protein kinase ,Mitogen-Activated Protein Kinase 3 ,MAP kinase kinase kinase ,Macrophages ,I-Kappa-B Kinase ,Articles ,Cell Biology ,MAP Kinase Kinase Kinases ,Molecular biology ,Recombinant Proteins ,I-kappa B Kinase ,Cell biology ,HEK293 Cells ,Gene Expression Regulation ,Plasmids ,Signal Transduction - Abstract
Tumor progression locus 2 (TPL-2) functions as a MEK-1/2 kinase, which is essential for Toll-like receptor 4 (TLR4) activation of extracellular signal-regulated kinase 1 and 2 (ERK-1/2) mitogen-activated protein (MAP) kinases in lipopolysaccharide (LPS)-stimulated macrophages and for inducing the production of the proinflammatory cytokines tumor necrosis factor and interleukin-1β. In unstimulated cells, association of TPL-2 with NF-κB1 p105 prevents TPL-2 phosphorylation of MEK-1/2. LPS stimulation of TPL-2 MEK-1/2 kinase activity requires TPL-2 release from p105. This is triggered by IκB kinase 2 (IKK-2) phosphorylation of the p105 PEST region, which promotes p105 ubiquitination and degradation by the proteasome. LPS activation of ERK-1/2 additionally requires transphosphorylation of TPL-2 on serine 400 in its C terminus, which controls TPL-2 signaling to ERK-1/2 independently of p105. However, the identity of the protein kinase responsible for TPL-2 serine 400 phosphorylation remained unknown. In the present study, we show that TPL-2 serine 400 phosphorylation is mediated by IKK2. The IKK complex therefore regulates two of the key regulatory steps required for TPL-2 activation of ERK-1/2, underlining the close linkage of ERK-1/2 MAP kinase activation to upregulation of NF-κB-dependent transcription.
- Published
- 2012
17. Arabidopsis thaliana histone deacetylase 14 (HDA14) is an α-tubulin deacetylase that associates with PP2A and enriches in the microtubule fraction with the putative histone acetyltransferase ELP3
- Author
-
Alison DeLong, Mhairi Nimick, Hue T. Tran, Greg B. G. Moorhead, Robert Gourlay, Nick Morrice, George W. Templeton, and R. Glen Uhrig
- Subjects
Histone deacetylase 5 ,HDAC11 ,Histone deacetylase 2 ,HDAC10 ,macromolecular substances ,Cell Biology ,Plant Science ,SAP30 ,Biology ,HDAC4 ,Biochemistry ,Histone H2A ,Genetics ,Histone deacetylase - Abstract
It is now emerging that many proteins are regulated by a variety of covalent modifications. Using microcystin-affinity chromatography we have purified multiple protein phosphatases and their associated proteins from Arabidopsis thaliana. One major protein purified was the histone deacetylase HDA14. We demonstrate that HDA14 can deacetylate α-tubulin, associates with α/β-tubulin and is retained on GTP/taxol-stabilized microtubules, at least in part, by direct association with the PP2A-A2 subunit. Like HDA14, the putative histone acetyltransferase ELP3 was purified on microcystin-Sepharose and is also enriched at microtubules, potentially functioning in opposition to HDA14 as the α-tubulin acetylating enzyme. Consistent with the likelihood of it having many substrates throughout the cell, we demonstrate that HDA14, ELP3 and the PP2A A-subunits A1, A2 and A3 all reside in both the nucleus and cytosol of the cell. The association of a histone deacetylase with PP2A suggests a direct link between protein phosphorylation and acetylation.
- Published
- 2012
18. Identification of the human testis protein phosphatase 1 interactome
- Author
-
Sandra Rebelo, Patricia T.W. Cohen, Sara C. Domingues, Luís Korrodi-Gregório, Sara L. C. Esteves, Odete A. B. da Cruz e Silva, Ana Paula Vintém, Margarida Fardilha, Edgar F. da Cruz e Silva, and Nick Morrice
- Subjects
Male ,DNA, Complementary ,Two-hybrid screening ,macromolecular substances ,Plasma protein binding ,Computational biology ,Biology ,environment and public health ,Biochemistry ,Interactome ,DNA-binding protein ,Gene Expression Regulation, Enzymologic ,Protein Phosphatase 1 ,Two-Hybrid System Techniques ,Chlorocebus aethiops ,Testis ,Animals ,Humans ,Protein Isoforms ,Protein phosphorylation ,Gene Library ,Pharmacology ,Genetics ,cDNA library ,fungi ,Protein phosphatase 1 ,COS Cells ,Sperm Motility ,Function (biology) ,Protein Binding ,Signal Transduction - Abstract
Protein phosphorylation is a critical regulatory mechanism in cellular signalling. To this end, PP1 is a major eukaryotic serine/threonine-specific phosphatase whose cellular functions, in turn, depend on complexes it forms with PP1 interacting proteins-PIPs. The importance of the testis/sperm-enriched variant, PP1γ2, in sperm motility and spermatogenesis has previously been shown. Given the key role of PIPs, it is imperative to identify the physiologically relevant PIPs in testis and sperm. Hence, we performed Yeast Two-Hybrid screens of a human testis cDNA library using as baits the different PP1 isoforms and also a proteomic approach aimed at identifying PP1γ2 binding proteins. To the best of our knowledge this is the largest data set of the human testis PP1 interactome. We report the identification of 77 proteins in human testis and 7 proteins in human sperm that bind PP1. The data obtained increased the known PP1 interactome by reporting 72 novel interactions. Confirmation of the interaction of PP1 with 5 different proteins was also further validated by co-immunoprecipitation or protein overlays. The data here presented provides important insights towards the function of these proteins and opens new possibilities for future research. In fact, such diversity in PP1 regulators makes them excellent targets for pharmacological intervention.
- Published
- 2011
19. Phosphoproteomic analysis reveals an intrinsic pathway for histone deacetylase 7 regulation that controls cytotoxic T lymphocyte function
- Author
-
Doreen A. Cantrell, Carmen Feijoo-Carnero, Nick Morrice, Jurgen Goebel, and María N. Navarro
- Subjects
Male ,Proteomics ,Immunology ,Green Fluorescent Proteins ,Molecular Sequence Data ,Receptors, Antigen, T-Cell ,chemical and pharmacologic phenomena ,Mice, Transgenic ,Biology ,Histone Deacetylases ,Mass Spectrometry ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cytosol ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Amino Acid Sequence ,Phosphorylation ,Transcription factor ,Cells, Cultured ,030304 developmental biology ,Oligonucleotide Array Sequence Analysis ,Regulation of gene expression ,Cell Nucleus ,0303 health sciences ,Microscopy, Confocal ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,T-cell receptor ,HDAC7 ,hemic and immune systems ,Phosphoproteins ,Molecular biology ,3. Good health ,Chromatin ,Mice, Inbred C57BL ,CTL ,030220 oncology & carcinogenesis ,Female ,Histone deacetylase ,Chromatography, Liquid ,Signal Transduction ,T-Lymphocytes, Cytotoxic - Abstract
Here we report an unbiased analysis of the cytotoxic T lymphocyte (CTL) serine-threonine phosphoproteome by high-resolution mass spectrometry. We identified approximately 2,000 phosphorylations in CTLs, of which approximately 450 were controlled by T cell antigen receptor (TCR) signaling. A significantly overrepresented group of molecules identified included transcription activators, corepressors and chromatin regulators. A focus on chromatin regulators showed that CTLs had high expression of the histone deacetylase HDAC7 but continually phosphorylated and exported this transcriptional repressor from the nucleus. Dephosphorylation of HDAC7 resulted in its accumulation in the nucleus and suppressed expression of genes encoding key cytokines, cytokine receptors and adhesion molecules that determine CTL function. Screening of the CTL phosphoproteome has thus identified intrinsic pathways of serine-threonine phosphorylation that target chromatin regulators and determine the CTL functional program.
- Published
- 2011
20. Checkpoint-dependent phosphorylation of Exo1 modulates the DNA damage response
- Author
-
Mikhajlo K. Zubko, Nick Morrice, Amanda Greenall, Hien-Ping Ngo, Isabelle Morin, and David Lydall
- Subjects
Proteomics ,Saccharomyces cerevisiae Proteins ,Genotype ,DNA damage ,DNA, Single-Stranded ,Cell Cycle Proteins ,Biology ,Protein Serine-Threonine Kinases ,medicine.disease_cause ,environment and public health ,Exo1 ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Article ,Fungal Proteins ,03 medical and health sciences ,chemistry.chemical_compound ,checkpoint ,medicine ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Mutation ,telomere ,General Immunology and Microbiology ,phosphorylation ,General Neuroscience ,030302 biochemistry & molecular biology ,Cell Cycle ,Temperature ,G2-M DNA damage checkpoint ,Cell cycle ,Molecular biology ,Telomere ,Cell biology ,enzymes and coenzymes (carbohydrates) ,Checkpoint Kinase 2 ,Exodeoxyribonucleases ,chemistry ,cdc13-1 ,DNA mismatch repair ,biological phenomena, cell phenomena, and immunity ,Genome, Fungal ,Replication fork processing ,DNA ,DNA Damage - Abstract
Exo1 is a nuclease involved in mismatch repair, DSB repair, stalled replication fork processing and in the DNA damage response triggered by dysfunctional telomeres. In budding yeast and mice, Exo1 creates single-stranded DNA (ssDNA) at uncapped telomeres. This ssDNA accumulation activates the checkpoint response resulting in cell cycle arrest. Here, we demonstrate that Exo1 is phosphorylated when telomeres are uncapped in cdc13-1 and yku70Delta yeast cells, and in response to the induction of DNA damage. After telomere uncapping, Exo1 phosphorylation depends on components of the checkpoint machinery such as Rad24, Rad17, Rad9, Rad53 and Mec1, but is largely independent of Chk1, Tel1 and Dun1. Serines S372, S567, S587 and S692 of Exo1 were identified as targets for phosphorylation. Furthermore, mutation of these Exo1 residues altered the DNA damage response to uncapped telomeres and camptothecin treatment, in a manner that suggests Exo1 phosphorylation inhibits its activity. We propose that Rad53-dependent Exo1 phosphorylation is involved in a negative feedback loop to limit ssDNA accumulation and DNA damage checkpoint activation.
- Published
- 2008
21. The IRAK-catalysed activation of the E3 ligase function of Pellino isoforms induces the Lys63-linked polyubiquitination of IRAK1
- Author
-
Emma Carrick, Nick Morrice, Hilary Smith, Mark Peggie, Mark Windheim, Philip Cohen, and Alban Ordureau
- Subjects
Protein subunit ,Transfection ,Biochemistry ,Catalysis ,Article ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,Humans ,Protein Isoforms ,Phosphorylation ,Molecular Biology ,Transcription factor ,030304 developmental biology ,Inflammation ,0303 health sciences ,biology ,Kinase ,Ubiquitination ,I-Kappa-B Kinase ,NF-kappa B p50 Subunit ,Cell Biology ,IRAK4 ,I-kappa B Kinase ,Ubiquitin ligase ,Enzyme Activation ,Interleukin-1 Receptor-Associated Kinases ,030220 oncology & carcinogenesis ,biology.protein ,Interleukin-1 - Abstract
The protein kinases IRAK [IL-1 (interleukin 1) receptor-associated kinase] 1 and 4 play key roles in a signalling pathway by which bacterial infection or IL-1 trigger the production of inflammatory mediators. In the present study, we demonstrate that IRAK1 and IRAK4 phosphorylate Pellino isoforms in vitro and that phosphorylation greatly enhances Pellino's E3 ubiquitin ligase activity. We show that, in vitro, Pellino 1 can combine with the E2 conjugating complex Ubc13 (ubiquitin-conjugating enzyme 13)-Uev1a (ubiquitin E2 variant 1a) to catalyse the formation of K63-pUb (Lys63-linked polyubiquitin) chains, with UbcH3 to catalyse the formation of K48-pUb chains and with UbcH4, UbcH5a or UbcH5b to catalyse the formation of pUb-chains linked mainly via Lys11 and Lys48 of ubiquitin. In IRAK1-/- cells, the co-transfection of DNA encoding wild-type IRAK1 and Pellino 2, but not inactive mutants of these proteins, induces the formation of K63-pUb-IRAK1 and its interaction with the NEMO [NF-kappaB (nuclear factor kappaB) essential modifier] regulatory subunit of the IKK (inhibitor of NF-kappaB kinase) complex, a K63-pUb-binding protein. These studies suggest that Pellino isoforms may be the E3 ubiquitin ligases that mediate the IL-1-stimulated formation of K63-pUb-IRAK1 in cells, which may contribute to the activation of IKKbeta and the transcription factor NF-kappaB, as well as other signalling pathways dependent on IRAK1/4.
- Published
- 2007
22. The nuclear PP1 interacting protein ZAP3 (ZAP) is a putative nucleoside kinase that complexes with SAM68, CIA, NF110/45, and HNRNP-G
- Author
-
Mark Glover, Angus I. Lamond, Laura Trinkle-Mulcahy, N. K. Bernstein, Greg B. G. Moorhead, Annegret Ulke-Lemée, Nick Morrice, and Steven G. Chaulk
- Subjects
Amino Acid Motifs ,Molecular Sequence Data ,Biophysics ,Cell Cycle Proteins ,Biology ,Biochemistry ,SH3 domain ,Analytical Chemistry ,MAP2K7 ,Protein Phosphatase 1 ,Animals ,Humans ,Amino Acid Sequence ,c-Raf ,Kinase activity ,Nuclear Factor 90 Proteins ,Protein kinase A ,Molecular Biology ,Conserved Sequence ,Adaptor Proteins, Signal Transducing ,Serine/threonine-specific protein kinase ,Binding Sites ,Heterogeneous-Nuclear Ribonucleoprotein Group F-H ,Phosphotransferases ,Nuclear Proteins ,RNA-Binding Proteins ,Rats ,DNA-Binding Proteins ,Repressor Proteins ,Phosphotransferases (Alcohol Group Acceptor) ,Protein Subunits ,Nucleoproteins ,Nuclear Factor 45 Protein ,Cyclin-dependent kinase 9 ,Rabbits ,Casein kinase 2 ,HeLa Cells ,Molecular Chaperones ,Protein Binding - Abstract
The targeting of protein kinases and phosphatases is fundamental to their roles as cellular regulators. The type one serine/threonine protein phosphatase (PP1) is enriched in the nucleus, yet few nuclear PP1 targeting subunits have been described and characterized. Here we show that the human protein, ZAP3 (also known as ZAP), is localized to the nucleus, that it is expressed in all mammalian tissues examined, and docks to PP1 through an RVRW motif located in its highly conserved carboxy-terminus. Proteomic analysis of a ZAP3 complex revealed that in addition to binding PP1, ZAP3 complexes with CIA (or nuclear receptor co-activator 5) and the RNA binding proteins hnRNP-G, SAM68 and NF110/45, but loses affinity for SAM68 and hnRNP-G upon digestion of endogenous nucleic acid. Bioinformatics has revealed that the conserved carboxy-terminus is orthologous to T4- and mammalian polynucleotide kinases with residues necessary for kinase activity maintained throughout evolution. Furthermore, the substrate binding pocket of uridine-cytidine kinase (or uridine kinase) has localized sequence similarity with ZAP3, suggesting uridine or cytidine as possible ZAP3 substrates. Most polynucleotide kinases have a phosphohydrolase domain in conjunction with their kinase domain. In ZAP3, although this domain is present, it now appears degenerate and functions to bind PP1 through an RVRW docking site located within the domain.
- Published
- 2007
23. A Phosphorylated Form of Mel-18 Targets the Ring1B Histone H2A Ubiquitin Ligase to Chromatin
- Author
-
Gordon Peters, Goedele N. Maertens, Neil Brockdorff, Nick Morrice, Haruhiko Koseki, Mitsuhiro Endoh, Sarah Elderkin, Kevin Hiom, and Donna L. Mallery
- Subjects
Macromolecular Substances ,Ubiquitin-Protein Ligases ,DNA Mutational Analysis ,Molecular Sequence Data ,macromolecular substances ,Biology ,Chromatin remodeling ,Histones ,Mice ,Histone H1 ,hemic and lymphatic diseases ,Histone H2A ,Animals ,Humans ,Histone code ,Amino Acid Sequence ,Histone octamer ,Phosphorylation ,Promoter Regions, Genetic ,Molecular Biology ,Cells, Cultured ,Embryonic Stem Cells ,Polycomb Repressive Complex 1 ,Ubiquitin ,Genes, Homeobox ,Zinc Fingers ,Cell Biology ,Chromatin ,Nucleosomes ,DNA-Binding Proteins ,Repressor Proteins ,Gene Expression Regulation ,Biochemistry ,Histone methyltransferase ,Chromatin immunoprecipitation - Abstract
Recent studies have shown that PRC1-like Polycomb repressor complexes monoubiquity-late chromatin on histone H2A at lysine residue 119. Here we have analyzed the function of the polycomb protein Mel-18. Using affinity-tagged human MEL-18, we identify a polycomb-like complex, melPRC1, containing the core PRC1 proteins, RING1/2, HPH2, and CBX8. We show that, in ES cells, melPRC1 can functionally substitute for other PRC1-like complexes in Hox gene repression. A reconstituted subcomplex containing only Ring1B and Mel-18 functions as an efficient ubiquitin E3 ligase. This complex ubiquitylates free histone substrates nonspecifically but is highly specific for histone H2A lysine 119 in the context of nucleosomes. Mutational analysis demonstrates that while Ring1B is required for E3 function, Mel-18 directs this activity to H2A lysine 119 in chromatin. Moreover, this substrate-targeting function of Mel-18 is dependent on its prior phosphorylation at multiple residues, providing a direct link between chromatin modification and cell signaling pathways.
- Published
- 2007
- Full Text
- View/download PDF
24. The DNA-Dependent Protein Kinase Catalytic Subunit Is Phosphorylated In Vivo on Threonine 3950, a Highly Conserved Amino Acid in the Protein Kinase Domain
- Author
-
Pauline Douglas, Ruiqiong Ye, Wesley D. Block, Katheryn Meek, Shikha Gupta, Xiaoping Cui, Yaping Yu, Susan P. Lees-Miller, Qi Ding, and Nick Morrice
- Subjects
Threonine ,Amino Acid Motifs ,Molecular Sequence Data ,DNA-Activated Protein Kinase ,Biology ,Mitogen-activated protein kinase kinase ,Protein Structure, Secondary ,Cell Line ,MAP2K7 ,DNA-Dependent Protein Kinase Catalytic Subunit ,Catalytic Domain ,Cell Line, Tumor ,Radiation, Ionizing ,Okadaic Acid ,Humans ,Amino Acid Sequence ,Enzyme Inhibitors ,Phosphorylation ,Molecular Biology ,Conserved Sequence ,DNA-PKcs ,Serine/threonine-specific protein kinase ,Sequence Homology, Amino Acid ,Autophosphorylation ,Articles ,Cell Biology ,Recombinant Proteins ,Protein Structure, Tertiary ,enzymes and coenzymes (carbohydrates) ,Biochemistry ,Cyclin-dependent kinase 9 ,Casein kinase 2 ,Protein Kinases - Abstract
The protein kinase activity of the DNA-dependent protein kinase (DNA-PK) is required for the repair of DNA double-strand breaks (DSBs) via the process of nonhomologous end joining (NHEJ). However, to date, the only target shown to be functionally relevant for the enzymatic role of DNA-PK in NHEJ is the large catalytic subunit DNA-PKcs itself. In vitro, autophosphorylation of DNA-PKcs induces kinase inactivation and dissociation of DNA-PKcs from the DNA end-binding component Ku70/Ku80. Phosphorylation within the two previously identified clusters of phosphorylation sites does not mediate inactivation of the assembled complex and only partially regulates kinase disassembly, suggesting that additional autophosphorylation sites may be important for DNA-PK function. Here, we show that DNA-PKcs contains a highly conserved amino acid (threonine 3950) in a region similar to the activation loop or t-loop found in the protein kinase domain of members of the typical eukaryotic protein kinase family. We demonstrate that threonine 3950 is an in vitro autophosphorylation site and that this residue, as well as other previously identified sites in the ABCDE cluster, is phosphorylated in vivo in irradiated cells. Moreover, we show that mutation of threonine 3950 to the phosphomimic aspartic acid abrogates V(D)J recombination and leads to radiation sensitivity. Together, these data suggest that threonine 3950 is a functionally important, DNA damage-inducible phosphorylation site and that phosphorylation of this site regulates the activity of DNA-PKcs.
- Published
- 2007
25. A Chaperone-Dependent GSK3β Transitional Intermediate Mediates Activation-Loop Autophosphorylation
- Author
-
Ross Kinstrie, Pamela A. Lochhead, Gary Sibbet, Nick Morrice, Vaughn Cleghon, and Teeara Rawjee
- Subjects
Protein Folding ,macromolecular substances ,Biology ,Cell Line ,MAP2K7 ,Glycogen Synthase Kinase 3 ,GSK-3 ,Animals ,Humans ,HSP90 Heat-Shock Proteins ,Enzyme Inhibitors ,Phosphorylation ,Kinase activity ,Molecular Biology ,MAPK14 ,Glycogen Synthase Kinase 3 beta ,Kinase ,Autophosphorylation ,Cell Biology ,Protein-Tyrosine Kinases ,Cell biology ,Enzyme Activation ,Gene Expression Regulation ,Biochemistry ,Tyrosine ,Tyrosine kinase ,Molecular Chaperones ,Signal Transduction - Abstract
Glycogen synthase kinase 3 (GSK3), a key component of the insulin and wnt signaling pathways, is unusual, as it is constitutively active and is inhibited in response to upstream signals. Kinase activity is thought to be increased by intramolecular phosphorylation of a tyrosine in the activation loop (Y216 in GSK3beta), whose timing and mechanism is undefined. We show that GSK3beta autophosphorylates Y216 as a chaperone-dependent transitional intermediate possessing intramolecular tyrosine kinase activity and displaying different sensitivity to small-molecule inhibitors compared to mature GSK3beta. After autophosphorylation, mature GSK3beta is then an intermolecular serine/threonine kinase no longer requiring a chaperone. This shows that autoactivating kinases have adopted different molecular mechanisms for autophosphorylation; and for kinases such as GSK3, inhibitors that affect only the transitional intermediate would be missed in conventional drug screens.
- Published
- 2006
26. FRAT1, a Substrate-specific Regulator of Glycogen Synthase Kinase-3 Activity, Is a Cellular Substrate of Protein Kinase A
- Author
-
Andrew West, Sheelagh Frame, Thilo Hagen, Darren A.E. Cross, Ainsley A. Culbert, Nick Morrice, and Alastair D. Reith
- Subjects
inorganic chemicals ,macromolecular substances ,Mitogen-activated protein kinase kinase ,environment and public health ,Biochemistry ,Cell Line ,Substrate Specificity ,MAP2K7 ,Glycogen Synthase Kinase 3 ,Proto-Oncogene Proteins ,Humans ,Protein phosphorylation ,ASK1 ,Phosphorylation ,Protein kinase A ,Molecular Biology ,Protein Kinase C ,beta Catenin ,Adaptor Proteins, Signal Transducing ,Cyclin-dependent kinase 1 ,biology ,MAP kinase kinase kinase ,Chemistry ,Cyclin-dependent kinase 2 ,Intracellular Signaling Peptides and Proteins ,Cell Biology ,Cyclic AMP-Dependent Protein Kinases ,Cell biology ,enzymes and coenzymes (carbohydrates) ,biology.protein ,bacteria - Abstract
FRAT1, like its Xenopus homolog glycogen synthase kinase-3 (GSK-3)-binding protein, is known to inhibit GSK-3-mediated phosphorylation of beta-catenin. It is currently unknown how FRAT-GSK-3-binding protein activity toward GSK-3 is regulated. FRAT1 has recently been shown to be a phosphoprotein in vivo; however, the responsible kinase(s) have not been determined. In this study, we identified Ser188 as a phosphorylated residue in FRAT1. The identity of the kinase that catalyzes Ser188 phosphorylation and the significance of this phosphorylation to FRAT1 function were investigated. Protein kinase A (PKA) was found to phosphorylate Ser188 in vitro as well as in intact cells. Importantly, activation of endogenous cAMP-coupled beta-adrenergic receptors with norepinephrine stimulated the phosphorylation of FRAT1 at Ser188. GSK-3 was also able to phosphorylate FRAT1 at Ser188 and other residues in vitro or when overexpressed in intact cells. In contrast, endogenous GSK-3 did not lead to significant FRAT1 phosphorylation in cells, suggesting that GSK-3 is not a major FRAT1 kinase in vivo. Phosphorylation of Ser188 by PKA inhibited the ability of FRAT1 to activate beta-catenin-dependent transcription. In conclusion, PKA phosphorylates FRAT1 in vitro as well as in intact cells and may play a role in regulating the inhibitory activity of FRAT1 toward GSK-3.
- Published
- 2006
27. DNA-PK autophosphorylation facilitates Artemis endonuclease activity
- Author
-
Caterina Marchetti, Pauline Douglas, Aaron A. Goodarzi, Nick Morrice, Penny A. Jeggo, Yaping Yu, Enriqueta Riballo, Sarah R. Walker, Susan P. Lees-Miller, Christine J. Härer, and Ruiqiong Ye
- Subjects
DNA Repair ,DCLRE1C ,Protein Conformation ,DNA repair ,DNA, Single-Stranded ,DNA-Activated Protein Kinase ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,chemistry.chemical_compound ,Endonuclease ,Catalytic Domain ,Serine ,Humans ,Phosphorylation ,Ku Autoantigen ,Molecular Biology ,Recombination, Genetic ,Nuclease ,General Immunology and Microbiology ,General Neuroscience ,Autophosphorylation ,DNA Helicases ,Nuclear Proteins ,DNA ,Endonucleases ,Molecular biology ,DNA-Binding Proteins ,Non-homologous end joining ,enzymes and coenzymes (carbohydrates) ,chemistry ,biology.protein - Abstract
The Artemis nuclease is defective in radiosensitive severe combined immunodeficiency patients and is required for the repair of a subset of ionising radiation induced DNA double-strand breaks (DSBs) in an ATM and DNA-PK dependent process. Here, we show that Artemis phosphorylation by ATM and DNA-PK in vitro is primarily attributable to S503, S516 and S645 and demonstrate ATM dependent phosphorylation at serine 645 in vivo. However, analysis of multisite phosphorylation mutants of Artemis demonstrates that Artemis phosphorylation is dispensable for endonuclease activity in vitro and for DSB repair and V(D)J recombination in vivo. Importantly, DNA-dependent protein kinase catalytic subunit (DNA-PKcs) autophosphorylation at the T2609-T2647 cluster, in the presence of Ku and target DNA, is required for Artemis-mediated endonuclease activity. Moreover, autophosphorylated DNA-PKcs stably associates with Ku-bound DNA with large single-stranded overhangs until overhang cleavage by Artemis. We propose that autophosphorylation triggers conformational changes in DNA-PK that enhance Artemis cleavage at single-strand to double-strand DNA junctions. These findings demonstrate that DNA-PK autophosphorylation regulates Artemis access to DNA ends, providing insight into the mechanism of Artemis mediated DNA end processing.
- Published
- 2006
28. Phosphorylation and 14-3-3 binding of Arabidopsis trehalose-phosphate synthase 5 in response to 2-deoxyglucose
- Author
-
Barry Hon Cheung Wong, Jean Harthill, Carol MacKintosh, Mark Peggie, Jonas Borch, Nick Morrice, and Sarah E. M. Meek
- Subjects
Threonine ,Recombinant Fusion Proteins ,Arabidopsis ,Mutation, Missense ,Plant Science ,AMP-Activated Protein Kinases ,Protein Serine-Threonine Kinases ,Deoxyglucose ,Biology ,Chromatography, Affinity ,Dephosphorylation ,AMP-Activated Protein Kinase Kinases ,Phenformin ,Multienzyme Complexes ,Serine ,Genetics ,Humans ,Immunoprecipitation ,Protein Isoforms ,Phosphorylation ,Protein kinase A ,Cells, Cultured ,Alanine ,Binding Sites ,ATP synthase ,Arabidopsis Proteins ,Kinase ,HEK 293 cells ,AMPK ,Cell Biology ,Protein-Serine-Threonine Kinases ,biology.organism_classification ,14-3-3 Proteins ,Biochemistry ,Glucosyltransferases ,biology.protein - Abstract
Udgivelsesdato: 2006-Jul Trehalose-6-phosphate is a 'sugar signal' that regulates plant metabolism and development. The Arabidopsis genome encodes trehalose-6-phosphate synthase (TPS) and trehalose-6-phosphatase (TPP) enzymes. It also encodes class II proteins (TPS isoforms 5-11) that contain both TPS-like and TPP-like domains, although whether these have enzymatic activity is unknown. In this paper, we show that TPS5, 6 and 7 are phosphoproteins that bind to 14-3-3 proteins, by using 14-3-3 affinity chromatography, 14-3-3 overlay assays, and by co-immunoprecipitating TPS5 and 14-3-3 isoforms from cell extracts. GST-TPS5 bound to 14-3-3s after in vitro phosphorylation at Ser22 and Thr49 by either mammalian AMP-activated protein kinase (AMPK) or partially purified plant Snf1-related protein kinase 1 (SnRK1s). Dephosphorylation of TPS5, or mutation of either Ser22 or Thr49, abolished binding to 14-3-3s. Ser22 and Thr49 are both conserved in TPS5, 7, 9 and 10. When GST-TPS5 was expressed in human HEK293 cells, Thr49 was phosphorylated in response to 2-deoxyglucose or phenformin, stimuli that activate the AMPK via the upstream kinase LKB1. 2-deoxyglucose stimulated Thr49 phosphorylation of endogenous TPS5 in Arabidopsis cells, whereas phenformin did not. Moreover, extractable SnRK1 activity was increased in Arabidopsis cells in response to 2-deoxyglucose. The plant kinase was inactivated by dephosphorylation and reactivated by phosphorylation with human LKB1, indicating that elements of the SnRK1/AMPK pathway are conserved in Arabidopsis and human cells. We hypothesize that coordinated phosphorylation and 14-3-3 binding of nitrate reductase (NR), 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (F2KP) and class II TPS isoforms mediate responses to signals that activate SnRK1.
- Published
- 2006
29. The PII Signal Transduction Protein of Arabidopsis thaliana Forms an Arginine-regulated Complex with Plastid N-Acetyl Glutamate Kinase
- Author
-
Yan M, Chen, Tony S, Ferrar, Elke M, Lohmeier-Vogel, Elke, Lohmeir-Vogel, Nick, Morrice, Yutaka, Mizuno, Byron, Berenger, Kenneth K S, Ng, Douglas G, Muench, and Greg B G, Moorhead
- Subjects
Chloroplasts ,Arginine ,PII Nitrogen Regulatory Proteins ,Arabidopsis ,Biology ,Biochemistry ,Affinity chromatography ,Arabidopsis thaliana ,Plastids ,Plastid ,Molecular Biology ,chemistry.chemical_classification ,Arabidopsis Proteins ,fungi ,Isothermal titration calorimetry ,Cell Biology ,Phosphotransferases (Carboxyl Group Acceptor) ,biology.organism_classification ,Enzyme assay ,Enzyme ,chemistry ,Multiprotein Complexes ,biology.protein ,Signal transduction ,Protein Binding ,Signal Transduction - Abstract
The PII proteins are key mediators of the cellular response to carbon and nitrogen status and are found in all domains of life. In eukaryotes, PII has only been identified in red algae and plants, and in these organisms, PII localizes to the plastid. PII proteins perform their role by assessing cellular carbon, nitrogen, and energy status and conferring this information to other proteins through protein-protein interaction. We have used affinity chromatography and mass spectrometry to identify the PII-binding proteins of Arabidopsis thaliana. The major PII-interacting protein is the chloroplast-localized enzyme N-acetyl glutamate kinase, which catalyzes the key regulatory step in the pathway to arginine biosynthesis. The interaction of PII with N-acetyl glutamate kinase was confirmed through pull-down, gel filtration, and isothermal titration calorimetry experiments, and binding was shown to be enhanced in the presence of the downstream product, arginine. Enzyme kinetic analysis showed that PII increases N-acetyl glutamate kinase activity slightly, but the primary function of binding is to relieve inhibition of enzyme activity by the pathway product, arginine. Knowing the identity of PII-binding proteins across a spectrum of photosynthetic and non-photosynthetic organisms provides a framework for a more complete understanding of the function of this highly conserved signaling protein.
- Published
- 2006
30. Substrate Specificity and Activity Regulation of Protein Kinase MELK
- Author
-
Hugo Ceulemans, Monique Beullens, Etienne Waelkens, Rita Derua, Nick Morrice, Sadia Vancauwenbergh, and Mathieu Bollen
- Subjects
Threonine ,Molecular Sequence Data ,Protein Array Analysis ,Buffers ,Protein Serine-Threonine Kinases ,Biology ,Ligands ,Peptide Mapping ,Biochemistry ,Gene Expression Regulation, Enzymologic ,Mass Spectrometry ,Cell Line ,Substrate Specificity ,Maternal embryonic leucine zipper kinase ,Enzyme activator ,Chlorocebus aethiops ,Serine ,Animals ,Humans ,Amino Acid Sequence ,RNA, Messenger ,Phosphorylation ,Binding site ,Protein kinase A ,Molecular Biology ,Ions ,Binding Sites ,Kinase ,Stem Cells ,Cell Cycle ,Alternative splicing ,Autophosphorylation ,Cell Biology ,Glutathione ,Recombinant Proteins ,Protein Structure, Tertiary ,Enzyme Activation ,Alternative Splicing ,Dithiothreitol ,Kinetics ,Gene Expression Regulation ,COS Cells ,Mutagenesis, Site-Directed ,Tyrosine ,Calcium ,Peptides ,Protein Binding - Abstract
Maternal embryonic leucine zipper kinase (MELK) is a protein Ser/Thr kinase that has been implicated in stem cell renewal, cell cycle progression, and pre-mRNA splicing, but its substrates and regulation are not yet known. We show here that MELK has a rather broad substrate specificity and does not appear to require a specific sequence surrounding its (auto)phosphorylation sites. We have mapped no less than 16 autophosphorylation sites including serines, threonines, and a tyrosine residue and show that the phosphorylation of Thr167 and Ser171 is required for the activation of MELK. The expression of MELK activity also requires reducing agents such as dithiothreitol or reduced glutathione. Furthermore, we show that MELK is a Ca2+-binding protein and is inhibited by physiological Ca2+ concentrations. The smallest MELK fragment that was still catalytically active comprises the N-terminal catalytic domain and the flanking ubiquitin-associated domain. A C-terminal fragment of MELK functions as an autoinhibitory domain. Our data show that the activity of MELK is regulated in a complex manner and offer new perspectives for the further elucidation of its biological function.
- Published
- 2005
31. Regulation of Microfilament Organization by Kaposi Sarcoma-associated Herpes Virus-cyclin·CDK6 Phosphorylation of Caldesmon
- Author
-
Axel Knebel, Nick Morrice, Maria Emanuela Cuomo, Georgina M. Platt, Sibylle Mittnacht, and Philip Cohen
- Subjects
Gene Expression Regulation, Viral ,Threonine ,Time Factors ,Microfilament ,Retinoblastoma Protein ,Biochemistry ,Catalysis ,Chromatography, Affinity ,Mass Spectrometry ,Substrate Specificity ,Mice ,Cyclin-dependent kinase ,Serine ,Animals ,Humans ,Cloning, Molecular ,Phosphorylation ,RNA, Small Interfering ,Molecular Biology ,Cytoskeleton ,Actin ,biology ,Kinase ,Sepharose ,Retinoblastoma protein ,Cyclin-Dependent Kinase 4 ,Cyclin-Dependent Kinase 6 ,Cell Biology ,Actin cytoskeleton ,Molecular biology ,Actins ,Recombinant Proteins ,Protein Structure, Tertiary ,Cell biology ,Caldesmon ,Microscopy, Fluorescence ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Herpesvirus 8, Human ,NIH 3T3 Cells ,biology.protein ,Calmodulin-Binding Proteins ,Electrophoresis, Polyacrylamide Gel ,Cyclin-dependent kinase 6 ,Peptides ,HeLa Cells ,Protein Binding - Abstract
Kaposi sarcoma-associated herpes virus (KSHV) encodes a D-like cyclin (K-cyclin) that is thought to contribute to the viral oncogenicity. K-cyclin activates cellular cyclin-dependent kinases (CDK) 4 and 6, generating enzymes with a substrate selectivity deviant from CDK4 and CDK6 activated by D-type cyclins, suggesting different biochemical and biological functions. Here we report the identification of the actin- and calmodulin-binding protein caldesmon (CALD1) as a novel K-cyclin.CDK substrate, which is not phosphorylated by D.CDK. CALD1 plays a central role in the regulation of microfilament organization, consequently controlling cell shape, adhesion, cytokinesis and motility. K-cyclin.CDK6 specifically phosphorylates four Ser/Thr sites in the human CALD1 carboxyl terminus, abolishing CALD1 binding to its effector protein, actin, and its regulator protein, calmodulin. CALD1 is hyperphosphorylated in cells following K-cyclin expression and in KSHV-transformed lymphoma cells. Moreover, expression of exogenous K-cyclin results in microfilament loss and changes in cell morphology; both effects are reliant on CDK catalysis and can be reversed by the expression of a phosphorylation defective CALD1. Together, these data strongly suggest that K-cyclin expression modulates the activity of caldesmon and through this the microfilament functions in cells. These results establish a novel link between KSHV infection and the regulation of the actin cytoskeleton.
- Published
- 2005
32. Identification of calcium-regulated heat-stable protein of 24 kDa (CRHSP24) as a physiological substrate for PKB and RSK using KESTREL
- Author
-
Philip Cohen, David G. Campbell, Nick Morrice, and Gillian C. Auld
- Subjects
Molecular Sequence Data ,Protein Serine-Threonine Kinases ,Mitogen-activated protein kinase kinase ,Ribosomal Protein S6 Kinases, 90-kDa ,Biochemistry ,Cell Line ,MAP2K7 ,Rats, Sprague-Dawley ,Proto-Oncogene Proteins ,Sequence Homology, Nucleic Acid ,Animals ,Humans ,ASK1 ,Amino Acid Sequence ,Phosphorylation ,Protein kinase A ,Molecular Biology ,Base Sequence ,MAP kinase kinase kinase ,biology ,Kinase ,Cyclin-dependent kinase 4 ,Cyclin-dependent kinase 2 ,Cell Biology ,Phosphoproteins ,Molecular biology ,Rats ,DNA-Binding Proteins ,Liver ,biology.protein ,Proto-Oncogene Proteins c-akt ,Transcription Factors ,Research Article - Abstract
A substrate for PKBa (protein kinase Bα) was detected in liver extracts, and was purified and identified as CRHSP24 (calcium-regulated heat-stable protein of apparent molecular mass 24 kDa). PKBa, as well as SGK1 (serum- and glucocorticoid-induced protein kinase 1) and RSK (p90 ribosomal S6 kinase), phosphorylated CRHSP24 stoichiometrically at Ser 5 2 in vitro and its brain-specific isoform PIPPin at the equivalent residue (Ser 5 8 ). CRHSP24 became phosphorylated at Ser 5 2 when HEK-293 (human embryonic kidney) cells were stimulated with IGF-1 (insulin-like growth factor-1) and this was prevented by inhibitors of PI3K (phosphoinositide 3-kinase), but not by rapamycin [an inhibitor of mTOR (mammalian target of rapamycin)] or PD 184352, an inhibitor of the classical MAPK (mitogen-activated protein kinase) cascade and hence the activation of RSK. IGF-1 induced a similarphosphorylation of CRHSP24 in ES (embryonic stem) cells from wild-type mice or mice that express the PDK I (3-phosphoinositide-dependent kinase I) mutant (PDK 1[L155E]) that activates PKBα normally, but cannot activate SGK. CRHSP24 also became phosphorylated at Ser 5 2 in response to EGF (epidermal growth factor) and this was prevented by blocking activation of both the classical MAPK cascade and the activation of PKBa, but not if just one of these pathways was inhibited. DYRK2 (dual-specificity tyrosine-phosphorylated and -regulated protein kinase 2) phosphorylated CRHSP24 at Ser 3 0 , Ser 3 2 and Ser 4 1 in vitro, and Ser 4 1 was identified as a site phosphorylated in cells. These and other results demonstrate that CRHSP24 is phosphorylated at Ser 5 2 by PKBa in response to IGF-1, at Ser 5 2 by PKBa and RSK in response to EGF, and at Ser 4 1 in the absence of IGF-1/EGF by a DYRK isofonn or another proline-directed protein kinase(s).
- Published
- 2005
33. The phosphorylation of CapZ-interacting protein (CapZIP) by stress-activated protein kinases triggers its dissociation from CapZ
- Author
-
Claire E. Eyers, Simon Arthur, Helen McNEILL, Axel Knebel, Nick Morrice, Ana Cuenda, and Philip Cohen
- Subjects
Molecular Sequence Data ,macromolecular substances ,Mitogen-activated protein kinase kinase ,Biology ,Biochemistry ,Substrate Specificity ,MAP2K7 ,Jurkat Cells ,Mice ,Phosphoserine ,Animals ,Humans ,Protein phosphorylation ,Amino Acid Sequence ,Cloning, Molecular ,Phosphorylation ,Protein kinase A ,Molecular Biology ,MAPK14 ,CapZ Actin Capping Protein ,Mice, Knockout ,Sequence Homology, Amino Acid ,Gene Expression Profiling ,Intracellular Signaling Peptides and Proteins ,Chromosome Mapping ,CapZ ,Cell Biology ,Molecular biology ,Actins ,Cell biology ,Molecular Weight ,Rabbits ,Mitogen-Activated Protein Kinases ,cGMP-dependent protein kinase ,Spleen ,Research Article ,Protein Binding ,Signal Transduction - Abstract
A protein expressed in immune cells and muscle was detected in muscle extracts as a substrate for several SAPKs (stress-activated protein kinases). It interacted specifically with the F-actin capping protein CapZ in splenocytes, and was therefore termed ‘CapZIP’ (CapZ-interacting protein). Human CapZIP was phosphorylated at Ser-179 and Ser-244 by MAPKAP-K2 (mitogen-activated protein kinase-activated protein kinase 2) or MAPKAP-K3 in vitro. Anisomycin induced the phosphorylation of CapZIP at Ser-179 in Jurkat cells, which was prevented by SB 203580, consistent with phosphorylation by MAPKAP-K2 and/or MAPKAP-K3. However, osmotic shock-induced phosphorylation of Ser-179 was unaffected by SB 203580. These and other results suggest that CapZIP is phosphorylated at Ser-179 in cells by MAPKAP-K2/MAPKAP-K3, and at least one other protein kinase. Stress-activated MAP kinase family members phosphorylated human CapZIP at many sites, including Ser-68, Ser-83, Ser-108 and Ser-216. Ser-108 became phosphorylated when Jurkat cells were exposed to osmotic shock, which was unaffected by SB 203580 and/or PD 184352, or in splenocytes from mice that do not express either SAPK3/p38γ or SAPK4/p38δ. Our results suggest that CapZIP may be phosphorylated by JNK (c-Jun N-terminal kinase), which phosphorylates CapZIP to >5 mol/mol within minutes in vitro. Osmotic shock or anisomycin triggered the dissociation of CapZIP from CapZ in Jurkat cells, suggesting that phosphorylation of CapZIP may regulate the ability of CapZ to remodel actin filament assembly in vivo.
- Published
- 2005
34. Protein Kinase A Regulates Caspase-9 Activation by Apaf-1 Downstream of Cytochrome c
- Author
-
Nick Morrice, Michelle Lickrish, Catherine Sampson, Paul Clarke, Lindsey A. Allan, and Morag C. Martin
- Subjects
Time Factors ,Xenopus ,DNA Mutational Analysis ,Molecular Sequence Data ,Apoptosis ,Mitogen-activated protein kinase kinase ,Biology ,Biochemistry ,MAP2K7 ,Cyclic AMP ,Serine ,Animals ,Humans ,Immunoprecipitation ,ASK1 ,Amino Acid Sequence ,c-Raf ,Phosphorylation ,Protein kinase A ,Molecular Biology ,Glutathione Transferase ,Cell-Free System ,Sequence Homology, Amino Acid ,MAP kinase kinase kinase ,Cytochromes c ,Proteins ,Cell Biology ,Cyclic AMP-Dependent Protein Kinases ,Caspase 9 ,Protein Structure, Tertiary ,Cell biology ,Enzyme Activation ,Apoptotic Protease-Activating Factor 1 ,Caspases ,Mutagenesis, Site-Directed ,Cyclin-dependent kinase 9 ,Apoptosome ,HeLa Cells ,Plasmids ,Signal Transduction - Abstract
The cyclic AMP signal transduction pathway modulates apoptosis in diverse cell types, although the mechanism is poorly understood. A critical component of the intrinsic apoptotic pathway is caspase-9, which is activated by Apaf-1 in the apoptosome, a large complex assembled in response to release of cytochrome c from mitochondria. Caspase-9 cleaves and activates effector caspases, predominantly caspase-3, resulting in the demise of the cell. Here we identified a distinct mechanism by which cyclic AMP regulates this apoptotic pathway through activation of protein kinase A. We show that protein kinase A inhibits activation of caspase-9 and caspase-3 downstream of cytochrome c in Xenopus egg extracts and in a human cell-free system. Protein kinase A directly phosphorylates human caspase-9 at serines 99, 183, and 195. However, mutational analysis demonstrated that phosphorylation at these sites is not required for the inhibitory effect of protein kinase A on caspase-9 activation. Importantly, protein kinase A inhibits cytochrome c-dependent recruitment of procaspase-9 to Apaf-1 but not activation of caspase-9 by a constitutively activated form of Apaf-1. These data indicate that extracellular signals that elevate cyclic AMP and activate protein kinase A may suppress apoptosis by inhibiting apoptosome formation downstream of cytochrome c release from mitochondria.
- Published
- 2005
35. Phosphorylation of a Distinct Structural Form of Phosphatidylinositol Transfer Protein α at Ser166 by Protein Kinase C Disrupts Receptor-mediated Phospholipase C Signaling by Inhibiting Delivery of Phosphatidylinositol to Membranes
- Author
-
Bruno Ségui, Neil Q. McDonald, Judith Murray-Rust, Gopal P. Sapkota, Nick Morrice, Alison Skippen, Shamshad Cockcroft, Clive P. Morgan, Victoria Allen-Baume, Banafshé Larijani, and Andrew Ball
- Subjects
Models, Molecular ,Threonine ,Time Factors ,Protein Conformation ,Biochemistry ,chemistry.chemical_compound ,Cytosol ,Serine ,Protein phosphorylation ,Phospholipid Transfer Proteins ,Phosphorylation ,Chromatography, High Pressure Liquid ,Protein Kinase C ,Chromatography ,Microscopy, Confocal ,Kinase ,Brain ,Recombinant Proteins ,Cell biology ,COS Cells ,Tetradecanoylphorbol Acetate ,Electrophoresis, Polyacrylamide Gel ,Signal Transduction ,Green Fluorescent Proteins ,HL-60 Cells ,Biology ,Transfection ,Animals ,Humans ,Phosphatidylinositol ,Molecular Biology ,Phosphatidylinositol transfer protein ,Protein kinase C ,Binding Sites ,Dose-Response Relationship, Drug ,Phospholipase C ,Cell Membrane ,Cell Biology ,Lipid Metabolism ,Protein Structure, Tertiary ,Rats ,chemistry ,Type C Phospholipases ,Mutation ,Mutagenesis, Site-Directed ,Isoelectric Focusing ,Peptides ,HeLa Cells ,Phosphatidylinositol transfer protein, alpha - Abstract
Phosphatidylinositol transfer protein alpha (PITPalpha) participates in the supply of phosphatidylinositol ( PI) required for many cellular events including phospholipase C (PLC) beta and gamma signaling by G-protein-coupled receptors and receptor-tyrosine kinases, respectively. Protein kinase C has been known to modulate PLC signaling by G-protein-coupled receptors and receptor-tyrosine kinases, although the molecular target has not been identified in most instances. In each case phorbol myristate acetate pretreatment of HL60, HeLa, and COS-7 cells abrogated PLC stimulation by the agonists formyl-Met-Leu-Phe, ATP, and epidermal growth factor, respectively. Here we show that phosphorylation of PITPalpha at Ser(166) resulted in inhibition of receptor-stimulated PLC activity. Ser(166) is localized in a small pocket between the 165 - 172 loop and the rest of the protein and was not solvent-accessible in either the PI- or phosphatidylcholine-loaded structures of PITPalpha. To allow phosphorylation at Ser(166), a distinct structural form is postulated, and mutation of Thr(59) to alanine shifted the equilibrium to this form, which could be resolved on native PAGE. The elution profile observed by size exclusion chromatography of phosphorylated PITPalpha from rat brain or in vitro phosphorylated PITPalpha demonstrated that phosphorylated PITPalpha is structurally distinct from the non-phosphorylated form. Phosphorylated PITPalpha was unable to deliver its PI cargo, although it could deliver phosphatidylcholine. We conclude that the PITPalpha structure has to relax to allow access to the Ser(166) site, and this may occur at the membrane surface where PI delivery is required for receptor-mediated PLC signaling.
- Published
- 2004
36. Proteomic Characterization of Protein Phosphatase Complexes of the Mammalian Nucleus
- Author
-
Annegret Ulke, Hue T. Tran, Christine Johannes, Greg B. G. Moorhead, and Nick Morrice
- Subjects
Male ,Proteomics ,Microcystins ,Protein subunit ,Phosphatase ,RNA-binding protein ,macromolecular substances ,Peptides, Cyclic ,environment and public health ,Biochemistry ,Mass Spectrometry ,Analytical Chemistry ,Protein Phosphatase 1 ,Phosphoprotein Phosphatases ,RNA Precursors ,Animals ,snRNP ,Phosphorylation ,Rats, Wistar ,Binding site ,Molecular Biology ,Cell Nucleus ,Binding Sites ,Chemistry ,Sepharose ,Binding protein ,Nuclear Proteins ,Protein phosphatase 1 ,Protein phosphatase 2 ,Chromatography, Agarose ,Rats ,Protein Binding - Abstract
Our knowledge of the serine/threonine protein phosphatases of the mammalian nucleus is limited compared with their cytosolic counterparts. Microcystin-Sepharose chromatography and mass spectrometry were utilized to affinity purify and identify protein phosphatase-associated proteins from isolated rat liver nuclei. Far Western analysis with labeled protein phosphatase 1 (PP1) showed that many more PP1 binding proteins exist in the nucleus than were previously demonstrated. Mass spectrometry confirmed the presence in the nucleus of the mammalian PP1 isoforms alpha1, alpha2, beta, and gamma1, plus the Aalpha and several of the B and B' subunits that are complexed to PP2A. Other proteins enriched on the microcystin matrix include the spliceosomal proteins known as the U2 snRNPs SAP145 and SAP155 and the U5 snRNPs p116 and p200, myosin heavy chain, and a nuclear PP1 myosin-targeting subunit related to M110. The putative RNA binding protein ZAP was also established as a nuclear PP1 binding protein using the criteria of co-purification with PP1 on microcystin-Sepharose, co-immunoprecipation, binding PP1 in an overlay assay, and presence of a putative PP1 binding site (KKRVRWAD). These results further support a key role for protein phosphatases in several nuclear functions, including the regulation of pre-mRNA splicing.
- Published
- 2004
37. Modulation of human insulin receptor substrate-1 tyrosine phosphorylation by protein kinase Cdelta
- Author
-
Robert S. Garofalo, Michael W. Greene, Nick Morrice, and Richard A. Roth
- Subjects
Insulin Receptor Substrate Proteins ,Molecular Sequence Data ,CHO Cells ,environment and public health ,Biochemistry ,Cell Line ,chemistry.chemical_compound ,Cricetinae ,Animals ,Humans ,Amino Acid Sequence ,Phosphorylation ,Tyrosine ,Kinase activity ,Protein kinase A ,Molecular Biology ,Protein Kinase C ,Protein kinase C ,biology ,Tyrosine phosphorylation ,Cell Biology ,Protein-Tyrosine Kinases ,Phosphoproteins ,Cell biology ,Isoenzymes ,Protein Kinase C-delta ,enzymes and coenzymes (carbohydrates) ,Insulin receptor ,chemistry ,Mutation ,biology.protein ,biological phenomena, cell phenomena, and immunity ,Peptides ,Research Article - Abstract
Non-esterified fatty acid (free fatty acid)-induced activation of the novel PKC (protein kinase C) isoenzymes PKCdelta and PKCtheta correlates with insulin resistance, including decreased insulin-stimulated IRS-1 (insulin receptor substrate-1) tyrosine phosphorylation and phosphoinositide 3-kinase activation, although the mechanism(s) for this resistance is not known. In the present study, we have explored the possibility of a novel PKC, PKCdelta, to modulate directly the ability of the insulin receptor kinase to tyrosine-phosphorylate IRS-1. We have found that expression of either constitutively active PKCdelta or wild-type PKCdelta followed by phorbol ester activation both inhibit insulin-stimulated IRS-1 tyrosine phosphorylation in vivo. Activated PKCdelta was also found to inhibit the IRS-1 tyrosine phosphorylation in vitro by purified insulin receptor using recombinant full-length human IRS-1 and a partial IRS-1-glutathione S-transferase-fusion protein as substrates. This inhibition in vitro was not observed with a non-IRS-1 substrate, indicating that it was not the result of a general decrease in the intrinsic kinase activity of the receptor. Consistent with the hypothesis that PKCdelta acts directly on IRS-1, we show that IRS-1 can be phosphorylated by PKCdelta on at least 18 sites. The importance of three of the PKCdelta phosphorylation sites in IRS-1 was shown in vitro by a 75-80% decrease in the incorporation of phosphate into an IRS-1 triple mutant in which Ser-307, Ser-323 and Ser-574 were replaced by Ala. More importantly, the mutation of these three sites completely abrogated the inhibitory effect of PKCdelta on IRS-1 tyrosine phosphorylation in vitro. These results indicate that PKCdelta modulates the ability of the insulin receptor to tyrosine-phosphorylate IRS-1 by direct phosphorylation of the IRS-1 molecule.
- Published
- 2004
38. WNK1, the kinase mutated in an inherited high-blood-pressure syndrome, is a novel PKB (protein kinase B)/Akt substrate
- Author
-
Sian Humphreys, Dario R. Alessi, Alberto C. Vitari, Maria Deak, Anne Phelan, Alan R. Prescott, Nick Morrice, and Barry Collins
- Subjects
Pseudohypoaldosteronism ,AKT1 ,P70-S6 Kinase 1 ,Protein Serine-Threonine Kinases ,Mitogen-activated protein kinase kinase ,Ribosomal Protein S6 Kinases, 90-kDa ,Biochemistry ,mTORC2 ,Catalysis ,Cell Line ,MAP2K7 ,Minor Histocompatibility Antigens ,WNK Lysine-Deficient Protein Kinase 1 ,Antibody Specificity ,Proto-Oncogene Proteins ,Humans ,Insulin-Like Growth Factor I ,Phosphorylation ,Molecular Biology ,Protein kinase C ,biology ,Akt/PKB signaling pathway ,Cyclin-dependent kinase 2 ,Intracellular Signaling Peptides and Proteins ,Syndrome ,Cell Biology ,Molecular biology ,Phosphothreonine ,Hypertension ,Mutation ,biology.protein ,Proto-Oncogene Proteins c-akt ,Research Article - Abstract
Recent evidence indicates that mutations in the gene encoding the WNK1 [with no K (lysine) protein kinase-1] results in an inherited hypertension syndrome called pseudohypoaldosteronism type II. The mechanisms by which WNK1 is regulated or the substrates it phosphorylates are currently unknown. We noticed that Thr-60 of WNK1, which lies N-terminal to the catalytic domain, is located within a PKB (protein kinase B) phosphorylation consensus sequence. We found that PKB phosphorylated WNK1 efficiently compared with known substrates, and both peptide map and mutational analysis revealed that the major PKB site of phosphorylation was Thr-60. Employing a phosphospecific Thr-60 WNK1 antibody, we demonstrated that IGF1 (insulin-like growth factor) stimulation of HEK-293 cells induced phosphorylation of endogenously expressed WNK1 at Thr-60. Consistent with PKB mediating this phosphorylation, inhibitors of PI 3-kinase (phosphoinositide 3-kinase; wortmannin and LY294002) but not inhibitors of mammalian target of rapamycin (rapamycin) or MEK1 (mitogen-activated protein kinase kinase-1) activation (PD184352), inhibited IGF1-induced phosphorylation of endogenous WNK1 at Thr-60. Moreover, IGF1-induced phosphorylation of endogenous WNK1 did not occur in PDK1−/− ES (embryonic stem) cells, in which PKB is not activated. In contrast, IGF1 still induced normal phosphorylation of WNK1 in PDK1L155E/L155E knock-in ES cells in which PKB, but not S6K (p70 ribosomal S6 kinase) or SGK1 (serum- and glucocorticoid-induced protein kinase 1), is activated. Our study provides strong pharmacological and genetic evidence that PKB mediates the phosphorylation of WNK1 at Thr-60 in vivo. We also performed experiments which suggest that the phosphorylation of WNK1 by PKB is not regulating its kinase activity or cellular localization directly. These results provide the first connection between the PI 3-kinase/PKB pathway and WNK1, suggesting a mechanism by which this pathway may influence blood pressure.
- Published
- 2004
39. Aurora B Regulates MCAK at the Mitotic Centromere
- Author
-
Karen Duncan, Paul D. Andrews, Linda Wordeman, Nick Morrice, Yulia Ovechkina, Michael Wagenbach, and Jason R. Swedlow
- Subjects
Fluorescent Antibody Technique ,Kinesins ,0302 clinical medicine ,Aurora Kinases ,Cricetinae ,Aurora Kinase B ,Electrophoresis, Gel, Two-Dimensional ,Phosphorylation ,RNA, Small Interfering ,Kinetochores ,Chromatography, High Pressure Liquid ,0303 health sciences ,Kinetochore ,Nocodazole ,Cell Cycle ,Cell biology ,Chromosome passenger complex ,030220 oncology & carcinogenesis ,Fluorescence Recovery After Photobleaching ,Paclitaxel ,Centromere ,Green Fluorescent Proteins ,Aurora B kinase ,Mitosis ,CHO Cells ,macromolecular substances ,In Vitro Techniques ,Protein Serine-Threonine Kinases ,Biology ,Transfection ,Models, Biological ,Chromosomes ,General Biochemistry, Genetics and Molecular Biology ,Kinetochore microtubule ,03 medical and health sciences ,Animals ,Humans ,Amino Acid Sequence ,Molecular Biology ,030304 developmental biology ,Cell Biology ,Antineoplastic Agents, Phytogenic ,Rats ,Spindle apparatus ,Luminescent Proteins ,Mutation ,Autoradiography ,HeLa Cells ,Developmental Biology - Abstract
Chromosome orientation and alignment within the mitotic spindle requires the Aurora B protein kinase and the mitotic centromere-associated kinesin (MCAK). Here, we report the regulation of MCAK by Aurora B. Aurora B inhibited MCAK's microtubule depolymerizing activity in vitro, and phospho-mimic (S/E) mutants of MCAK inhibited depolymerization in vivo. Expression of either MCAK (S/E) or MCAK (S/A) mutants increased the frequency of syntelic microtubule-kinetochore attachments and mono-oriented chromosomes. MCAK phosphorylation also regulates MCAK localization: the MCAK (S/E) mutant frequently localized to the inner centromere while the (S/A) mutant concentrated at kinetochores. We also detected two different binding sites for MCAK using FRAP analysis of the different MCAK mutants. Moreover, disruption of Aurora B function by expression of a kinase-dead mutant or RNAi prevented centromeric targeting of MCAK. These results link Aurora B activity to MCAK function, with Aurora B regulating MCAK's activity and its localization at the centromere and kinetochore.
- Published
- 2004
40. Inhibition of caspase-9 through phosphorylation at Thr 125 by ERK MAPK
- Author
-
Shalini Pathak, Nick Morrice, Suzanne C. Brady, Gareth Magee, Lindsey A. Allan, and Paul Clarke
- Subjects
Threonine ,MAPK/ERK pathway ,Cell Survival ,Pyridines ,Recombinant Fusion Proteins ,Molecular Sequence Data ,MAP Kinase Kinase 1 ,Apoptosis ,Cytochrome c Group ,Protein Serine-Threonine Kinases ,Mice ,Epidermal growth factor ,medicine ,Animals ,Humans ,Staurosporine ,Enzyme Inhibitors ,Phosphorylation ,Protein kinase A ,Tissue homeostasis ,Caspase ,Mitogen-Activated Protein Kinase Kinases ,Base Sequence ,Epidermal Growth Factor ,biology ,Caspase 3 ,Kinase ,3T3 Cells ,Cell Biology ,Caspase 9 ,Cell biology ,Cell Transformation, Neoplastic ,Eukaryotic Cells ,Caspases ,Cancer research ,biology.protein ,Mitogen-Activated Protein Kinases ,HeLa Cells ,Signal Transduction ,medicine.drug - Abstract
Many pro-apoptotic signals activate caspase-9, an initiator protease that activates caspase-3 and downstream caspases to initiate cellular destruction1. However, survival signals can impinge on this pathway and suppress apoptosis. Activation of the Ras–Raf–MEK–ERK mitogen-activated protein kinase (MAPK) pathway is associated with protection of cells from apoptosis and inhibition of caspase-3 activation2,3,4,5, although the targets are unknown. Here, we show that the ERK MAPK pathway inhibits caspase-9 activity by direct phosphorylation. In mammalian cell extracts, cytochrome c-induced activation of caspases-9 and -3 requires okadaic-acid-sensitive protein phosphatase activity. The opposing protein kinase activity is overcome by treatment with the broad-specificity kinase inhibitor staurosporine or with inhibitors of MEK1/2. Caspase-9 is phosphorylated at Thr 125, a conserved MAPK consensus site targeted by ERK2 in vitro, in a MEK-dependent manner in cells stimulated with epidermal growth factor (EGF) or 12-O-tetradecanoylphorbol-13-acetate (TPA). Phosphorylation at Thr 125 is sufficient to block caspase-9 processing and subsequent caspase-3 activation. We suggest that phosphorylation and inhibition of caspase-9 by ERK promotes cell survival during development and tissue homeostasis. This mechanism may also contribute to tumorigenesis when the ERK MAPK pathway is constitutively activated.
- Published
- 2003
41. Protein phosphatase 4 interacts with the Survival of Motor Neurons complex and enhances the temporal localisation of snRNPs
- Author
-
Judith E. Sleeman, Angus I. Lamond, Patricia T.W. Cohen, Amanda Philp, Nick Morrice, Graeme K. Carnegie, C. James Hastie, and Mark Peggie
- Subjects
DNA, Complementary ,Time Factors ,Protein subunit ,Phosphatase ,Coiled Bodies ,Nerve Tissue Proteins ,RNA-binding protein ,Biology ,Transfection ,Cell Line ,DEAD-box RNA Helicases ,Minor Histocompatibility Antigens ,Muscular Atrophy, Spinal ,Epitopes ,Protein structure ,DEAD Box Protein 20 ,SMN complex ,SMN Complex Proteins ,Phosphoprotein Phosphatases ,Humans ,Tissue Distribution ,snRNP ,Cyclic AMP Response Element-Binding Protein ,In Situ Hybridization, Fluorescence ,Cell Nucleus ,Nuclear Proteins ,RNA-Binding Proteins ,Cell Biology ,Protein phosphatase 2 ,Blotting, Northern ,Ribonucleoproteins, Small Nuclear ,Precipitin Tests ,Protein Structure, Tertiary ,nervous system diseases ,Microscopy, Fluorescence ,Biochemistry ,Chromosomes, Human, Pair 5 ,RNA ,Electrophoresis, Polyacrylamide Gel ,Chromosomes, Human, Pair 3 ,Dimerization ,RNA Helicases ,HeLa Cells ,Plasmids ,Protein Binding - Abstract
Protein phosphatase 4 (PPP4) is a ubiquitous essential protein serine/threonine phosphatase found in higher eukaryotes. Coordinate variation of the levels of the catalytic subunit (PPP4c) and the regulatory subunit (R2)suggests that PPP4c and R2 form a heterodimeric core to which other regulatory subunits bind. Two proteins that specifically co-purify with Flag-epitope-tagged R2 expressed in HEK-293 cells were identified as Gemin3 and Gemin4. These two proteins have been identified previously as components of the Survival of Motor Neurons (SMN) protein complex, which is functionally defective in the hereditary disorder spinal muscular atrophy. Immuno-sedimentation of the epitope-tagged SMN protein complex from HeLa cells expressing CFP-SMN showed that the SMN protein interacts, as previously reported, with Gemin2 (SIP1), Gemin3 and Gemin4 and in addition associates with PPP4c. The SMN complex has been implicated in the assembly and maturation of small nuclear ribonucleoproteins (snRNPs). Expression of GFP-R2–PPP4c in HeLa cells enhances the temporal localisation of newly formed snRNPs, which is consistent with an association of R2-PPP4c with the SMN protein complex.
- Published
- 2003
42. Purification of a plant nucleotide pyrophosphatase as a protein that interferes with nitrate reductase and glutamine synthetase assays
- Author
-
Catherine S. Smith, Dave Bridges, Carol MacKintosh, Nick Morrice, Greg Moorhead, Sarah E. M. Meek, and Pauline Douglas
- Subjects
Molecular Sequence Data ,Brassica ,medicine.disease_cause ,Nitrate reductase ,Nitrate Reductase ,Biochemistry ,Nudix hydrolase ,Adenosine Triphosphate ,Glutamate-Ammonia Ligase ,Nitrate Reductases ,Glutamine synthetase ,medicine ,Nucleotide ,Amino Acid Sequence ,Enzyme Inhibitors ,Pyrophosphatases ,Escherichia coli ,Plant Proteins ,chemistry.chemical_classification ,Oxidase test ,biology ,NAD ,biology.organism_classification ,Molecular biology ,Adenosine Monophosphate ,In vitro ,chemistry ,Flavin-Adenine Dinucleotide ,Spinach ,Biological Assay ,Sequence Alignment - Abstract
An activity that inhibited both glutamine synthetase (GS) and nitrate reductase (NR) was highly purified from cauliflower (Brassica oleracea var. botrytis) extracts. The final preparation contained an acyl-CoA oxidase and a second protein of the plant nucleotide pyrophosphatase family. This preparation hydrolysed NADH, ATP and FAD to generate AMP and was inhibited by fluoride, Cu2+, Zn2+ and Ni2+. The purified fraction had no effect on the activity of NR when reduced methylviologen was used as electron donor instead of NADH; and inhibited the oxidation of NADH by both spinach NR and an Escherichia coli extract in a time-dependent manner. The apparent inhibition of GS and NR and the ability of ATP and AMP to relieve the inhibition of NR can therefore be explained by hydrolysis of nucleotide substrates by the nucleotide pyrophosphatase. We have no evidence that the nucleotide pyrophosphatase is a specific physiological regulator of NR and GS, but suggest that nucleotide pyrophosphatase activity may underlie some confusion in the literature about the effects of nucleotides and protein factors on NR and GS in vitro.
- Published
- 2003
43. Inhibition of SAPK2a/p38 prevents hnRNP A0 phosphorylation by MAPKAP-K2 and its interaction with cytokine mRNAs
- Author
-
Nick Morrice, David G. Campbell, Matthias Gaestel, Simon Rousseau, Mark Peggie, and Philip Cohen
- Subjects
Lipopolysaccharide ,Pyridines ,SB 203580 ,medicine.medical_treatment ,p38 mitogen-activated protein kinases ,Chemokine CXCL2 ,Protein Serine-Threonine Kinases ,Biology ,p38 Mitogen-Activated Protein Kinases ,Heterogeneous-Nuclear Ribonucleoproteins ,General Biochemistry, Genetics and Molecular Biology ,Mice ,chemistry.chemical_compound ,medicine ,Animals ,RNA, Messenger ,Enzyme Inhibitors ,Phosphorylation ,Molecular Biology ,AU-rich element ,Messenger RNA ,General Immunology and Microbiology ,Tumor Necrosis Factor-alpha ,Monokines ,General Neuroscience ,Imidazoles ,Intracellular Signaling Peptides and Proteins ,Articles ,Molecular biology ,In vitro ,Cytokine ,chemistry ,Mitogen-Activated Protein Kinases - Abstract
Lipopolysaccharide (LPS) stimulates production of inflammatory mediators, partly by stabilizing [interleukin-6 (IL-6), cyclooxygenase 2 (COX-2)] and/or stimulating translation [tumour necrosis factor-alpha (TNF-alpha)] of their mRNAs. Such regulation depends on AU-rich elements (AREs) within the 3'-untranslated regions and is partially suppressed by SB 203580 (which inhibits SAPK2a/p38). The LPS-induced production of TNF-alpha and IL-6 is suppressed in MAPKAP-K2-deficient mice (a kinase activated by SAPK2a/p38). Here, we identify 18 macrophage proteins that bind to AREs and show that hnRNP A0 is a major substrate for MAPKAP-K2 in this fraction. MAPKAP-K2 phosphorylated hnRNP A0 at Ser84 in vitro and this residue became phosphorylated in LPS-stimulated cells. Phosphorylation was prevented by SB 203580 and suppressed in macrophages derived from MAPKAP-K2-deficient mice. The mRNAs encoding TNF-alpha, COX-2 and macrophage inflammatory protein-2 (MIP-2) bound to hnRNP A0 in LPS-stimulated macrophages, an interaction prevented by SB 203580. The LPS-induced stabilization of MIP-2 mRNA and production of MIP-2 protein were abolished when macrophages were incubated with SB 203580 plus PD 184352 (which inhibits the classical MAP kinase cascade). Our data suggest that LPS-induced binding of hnRNP A0 to AREs may contribute to the post-transcriptional regulation of specific mRNAs.
- Published
- 2002
44. Stress-induced regulation of eukaryotic elongation factor 2 kinase by SB 203580-sensitive and −insensitive pathways
- Author
-
Axel Knebel, Claire E. Haydon, Nick Morrice, and Philip Cohen
- Subjects
Elongation Factor 2 Kinase ,inorganic chemicals ,Pyridines ,Molecular Sequence Data ,macromolecular substances ,Protein Serine-Threonine Kinases ,Biology ,Mitogen-activated protein kinase kinase ,p38 Mitogen-Activated Protein Kinases ,environment and public health ,Biochemistry ,MAP2K7 ,Mitogen-Activated Protein Kinase 13 ,chemistry.chemical_compound ,Mitogen-Activated Protein Kinase 11 ,Peptide Elongation Factor 2 ,Stress, Physiological ,Serine ,Tumor Cells, Cultured ,Humans ,ASK1 ,Amino Acid Sequence ,Enzyme Inhibitors ,Phosphorylation ,Molecular Biology ,Anisomycin ,MAPK14 ,Dose-Response Relationship, Drug ,MAP kinase kinase kinase ,Tumor Necrosis Factor-alpha ,Cyclin-dependent kinase 2 ,Imidazoles ,Intracellular Signaling Peptides and Proteins ,Cell Biology ,Molecular biology ,enzymes and coenzymes (carbohydrates) ,chemistry ,Calcium-Calmodulin-Dependent Protein Kinases ,biology.protein ,bacteria ,Cyclin-dependent kinase 9 ,Mitogen-Activated Protein Kinases ,Research Article ,Signal Transduction - Abstract
Eukaryotic elongation factor 2 (eEF2) kinase, the enzyme that inactivates eEF2, is controlled by phosphorylation. Previous work showed that stress-activated protein kinase 4 (SAPK4, also called p38delta) inhibits eEF2 kinase in vitro by phosphorylating Ser-359, while ribosomal protein S6 kinases inhibit eEF2 kinase by phosphorylating Ser-366 [Knebel, Morrice and Cohen (2001) EMBO J. 20, 4360-4369; Wang, Li, Williams, Terada, Alessi and Proud (2001) EMBO J. 20, 4370-4379]. In the present study we have examined the effects of the protein synthesis inhibitor anisomycin and tumour necrosis factor-alpha (TNF-alpha) on the phosphorylation of eEF2 kinase. We demonstrate that Ser-359, Ser-366 and two novel sites (Ser-377 and Ser-396) are all phosphorylated in human epithelial KB cells, but only the phosphorylation of Ser-359 and Ser-377 increases in response to these agonists and correlates with the dephosphorylation (activation) of eEF2. Ser-377 is probably a substrate of MAPKAP-K2/K3 (mitogen-activated protein kinase-activated protein kinase 2/kinase 3) in cells, because eEF2 kinase is phosphorylated efficiently by these protein kinases in vitro and phosphorylation of this site, induced by TNF-alpha and low (but not high) concentrations of anisomycin, is prevented by SB 203580, which inhibits SAPK2a/p38, their "upstream" activator. The phosphorylation of Ser-359 induced by high concentrations of anisomycin is probably catalysed by SAPK4/p38delta in cells, because no other stress-activated, proline-directed protein kinase tested phosphorylates this site in vitro and phosphorylation is insensitive to SB 203580. Interestingly, the phosphorylation of Ser-359 induced by TNF-alpha or low concentrations of anisomycin is suppressed by SB 203580, indicating that phosphorylation is also mediated by a novel pathway. Since the phosphorylation of Ser-377 does not inhibit eEF2 kinase in vitro, our results suggest that anisomycin or TNF-alpha inhibit eEF2 kinase via the phosphorylation of Ser-359.
- Published
- 2002
45. Molecular Basis for the Substrate Specificity of NIMA-related Kinase-6 (NEK6)
- Author
-
Maria Deak, Liying Dong, Mike Schutkowski, C. James Hastie, Jose M. Lizcano, Nick Morrice, Dario R. Alessi, Ulf Reimer, and Agnieszka Kieloch
- Subjects
MAP kinase kinase kinase ,biology ,Cyclin-dependent kinase 2 ,P70-S6 Kinase 1 ,Cell Biology ,Mitogen-activated protein kinase kinase ,Biochemistry ,MAP2K7 ,biology.protein ,ASK1 ,Cyclin-dependent kinase 9 ,Molecular Biology ,cGMP-dependent protein kinase - Abstract
The AGC family of protein kinases, which includes isoforms of protein kinase B (also known as Akt), ribosomal S6 protein kinase (S6K), and serum- and glucocorticoid-induced protein kinase (SGK) are activated in response to many extracellular signals and play key roles in regulating diverse cellular processes. They are activated by the phosphorylation of the T loop of their kinase domain by the 3-phosphoinositide-dependent protein kinase-1 and by phosphorylation of a residue located C-terminal to the kinase domain in a region termed the hydrophobic motif. Recent work has implicated the NIMA (never in mitosis, gene A)-related kinase-6 (NEK6) as the enzyme that phosphorylates the hydrophobic motif of S6K1 in vivo. Here we demonstrate that in addition to phosphorylating S6K1 and SGK1 at their hydrophobic motif, NEK6 also phosphorylates S6K1 at two other sites and phosphorylates SGK1 at one other site in vitro. Employing the Jerini pepSTAR method in combination with kinetic analysis of phosphorylation of variant peptides, we establish the key substrate specificity determinants for NEK6. Our analysis indicates that NEK6 has a strong preference for Leu 3 residues N-terminal to the site of phosphorylation. Its mutation to either Ile or Val severely reduced the efficacy with which NEK6-phosphorylated peptide substrates, and moreover, mutation of the equivalent Leu residue in S6K1 or SGK1 prevented phosphorylation of their hydrophobic motifs by NEK6 in vitro. However, these mutants of S6K1 or SGK1 still became phosphorylated at their hydrophobic motif following insulin-like growth factor-1 stimulation of transfected 293 cells. This study provides the first description of the basis for the substrate specificity of NEK6 and indicates that NEK6 is unlikely to be responsible for the IGF1-induced phosphorylation of the hydrophobic motif of S6K, SGK, and protein kinase B isoforms in vivo.
- Published
- 2002
46. Identification of a Phosphorylation Site on Skeletal Muscle Myosin Light Chain Kinase That Becomes Phosphorylated during Muscle Contraction
- Author
-
Matthias Gaestel, Philip Cohen, Peter Watt, Claire E. Haydon, Axel Knebel, and Nick Morrice
- Subjects
inorganic chemicals ,Myosin light-chain kinase ,Tyrosine 3-Monooxygenase ,Arginine ,Calmodulin ,Molecular Sequence Data ,Biophysics ,macromolecular substances ,Protein Serine-Threonine Kinases ,Biology ,p38 Mitogen-Activated Protein Kinases ,Biochemistry ,Substrate Specificity ,Serine ,medicine ,Animals ,Protein phosphorylation ,Amino Acid Sequence ,Phosphorylation ,Muscle, Skeletal ,Myosin-Light-Chain Kinase ,Molecular Biology ,Histidine ,Intracellular Signaling Peptides and Proteins ,Skeletal muscle ,Actins ,Electric Stimulation ,Hindlimb ,Cell biology ,Isoenzymes ,enzymes and coenzymes (carbohydrates) ,medicine.anatomical_structure ,14-3-3 Proteins ,Calcium-Calmodulin-Dependent Protein Kinases ,biology.protein ,Rabbits ,Mitogen-Activated Protein Kinases ,medicine.symptom ,Muscle Contraction ,Protein Binding ,Muscle contraction - Abstract
A protein phosphorylated efficiently in vitro by MAP kinase-activated protein kinase-2 (MAPKAP-K2) was purified from skeletal muscle extracts and identified as the calcium/calmodulin-dependent myosin light chain kinase (MLCK). The phosphorylation site was mapped to Ser 161 , a residue shown previously to be autophosphorylated by MLCK. The residue equivalent to Ser 161 became phosphorylated in vivo when rat hindlimbs were stimulated electrically. However, phosphorylation was triggered within seconds, whereas activation of MAPKAP-K2 required several minutes. Moreover, contraction-induced Ser 161 phosphorylation was similar in wild-type or MAPKAP-K2−/− mice. These results indicate that contraction-induced phosphorylation is probably catalyzed by MLCK and not MAPKAP-K2. Ser 161 phosphorylation induced the binding of MLCK to 14-3-3 proteins, but did not detectably affect the kinetic properties of MLCK. The sequence surrounding Ser 161 is unusual in that residue 158 is histidine. Previously, an arginine located three residues N-terminal to the site of phosphorylation was thought to be critical for the specificity of MAPKAP-K2.
- Published
- 2002
47. Chk1 protects against chromatin bridges by constitutively phosphorylating BLM serine 502 to inhibit BLM degradation
- Author
-
Eleni Petsalaki, George Zachos, Nick Morrice, and Maria Dandoulaki
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Amino Acid Motifs ,Biology ,environment and public health ,Chromatin bridge ,Cell Line, Tumor ,Serine ,Humans ,CHEK1 ,Phosphorylation ,Mitosis ,Anaphase ,RecQ Helicases ,urogenital system ,nutritional and metabolic diseases ,Cell Biology ,Cullin Proteins ,Molecular biology ,Chromatin ,Cell biology ,enzymes and coenzymes (carbohydrates) ,Checkpoint Kinase 1 ,Proteolysis ,biology.protein ,Chromosome breakage ,Protein Kinases ,Cullin ,Protein Binding - Abstract
Chromatin bridges represent incompletely segregated chromosomal DNA connecting the anaphase poles and can result in chromosome breakage. The Bloom's syndrome protein helicase (BLM, also known as BLMH) suppresses formation of chromatin bridges. Here, we show that cells deficient in checkpoint kinase 1 (Chk1, also known as CHEK1) exhibit higher frequency of chromatin bridges and reduced BLM protein levels compared to controls. Chk1 inhibition leads to BLM ubiquitylation and proteasomal degradation during interphase. Furthermore, Chk1 constitutively phosphorylates human BLM at serine 502 (S502) and phosphorylated BLM localises to chromatin bridges. Mutation of S502 to a non-phosphorylatable alanine residue (BLM-S502A) reduces the stability of BLM, whereas expression of a phospho-mimicking BLM-S502D, in which S502 is mutated to aspartic acid, stabilises BLM and prevents chromatin bridges in Chk1-deficient cells. In addition, wild-type but not BLM-S502D associates with cullin 3, and cullin 3 depletion rescues BLM accumulation and localisation to chromatin bridges after Chk1 inhibition. We propose that Chk1 phosphorylates BLM-S502 to inhibit cullin-3-mediated BLM degradation during interphase. These results suggest that Chk1 prevents deleterious anaphase bridges by stabilising BLM.
- Published
- 2014
48. 5′-AMP-activated Protein Kinase Phosphorylates IRS-1 on Ser-789 in Mouse C2C12 Myotubes in Response to 5-Aminoimidazole-4-carboxamide Riboside
- Author
-
D. Grahame Hardie, Hans Tornqvist, Nick Morrice, and Søren Nyboe Jakobsen
- Subjects
medicine.medical_specialty ,Time Factors ,Blotting, Western ,AMP-Activated Protein Kinases ,Protein Serine-Threonine Kinases ,Biochemistry ,5'-AMP-Activated Protein Kinase ,Mice ,Phosphatidylinositol 3-Kinases ,chemistry.chemical_compound ,Multienzyme Complexes ,Internal medicine ,Serine ,medicine ,Animals ,Humans ,Insulin ,Phosphatidylinositol ,Phosphorylation ,Muscle, Skeletal ,Protein kinase A ,Molecular Biology ,Cells, Cultured ,Binding Sites ,Dose-Response Relationship, Drug ,biology ,Myogenesis ,Myocardium ,Glucose transporter ,AMPK ,Biological Transport ,Cell Biology ,Aminoimidazole Carboxamide ,Phosphoproteins ,Recombinant Proteins ,Enzyme Activation ,Insulin receptor ,Glucose ,Endocrinology ,chemistry ,Insulin Receptor Substrate Proteins ,biology.protein ,Electrophoresis, Polyacrylamide Gel ,Ribonucleosides ,Peptides ,Protein Binding ,Signal Transduction ,Subcellular Fractions - Abstract
Exercise is known to increase insulin sensitivity and is an effective form of treatment for the hyperglycemia observed in type 2 diabetes. Activation of 5'-AMP-activated protein kinase (AMPK) by 5-aminoimidazole-4-carboxamide riboside (AICAR), exercise, or electrically stimulated contraction leads to increased glucose transport in skeletal muscle. Here we report the first evidence of a direct interaction between AMPK and the most upstream component of the insulin-signaling cascade, insulin receptor substrate-1 (IRS-1). We find that AMPK rapidly phosphorylates IRS-1 on Ser-789 in cell-free assays as well as in mouse C2C12 myotubes incubated with AICAR. In the C2C12 myotubes activation of AMPK by AICAR matched the phosphorylation of IRS-1 on Ser-789. This phosphorylation correlates with a 65% increase in insulin-stimulated IRS-1-associated phosphatidylinositol 3-kinase activity in C2C12 myotubes preincubated with AICAR. The binding of phosphatidylinositol 3-kinase to IRS-1 was not affected by AICAR. These results demonstrate the existence of an interaction between AMPK and early insulin signaling that could be of importance to our understanding of the potentiating effects of exercise on insulin signaling.
- Published
- 2001
49. A novel method to identify protein kinase substrates: eEF2 kinase is phosphorylated and inhibited by SAPK4/p38delta
- Author
-
Axel Knebel, Nick Morrice, and Philip Cohen
- Subjects
Elongation Factor 2 Kinase ,MAP Kinase Kinase 4 ,Molecular Sequence Data ,MAP Kinase Kinase 1 ,P70-S6 Kinase 1 ,MAP Kinase Kinase 6 ,Protein Serine-Threonine Kinases ,Mitogen-activated protein kinase kinase ,p38 Mitogen-Activated Protein Kinases ,Article ,General Biochemistry, Genetics and Molecular Biology ,Substrate Specificity ,MAP2K7 ,Mitogen-Activated Protein Kinase 13 ,Mitogen-Activated Protein Kinase 12 ,Humans ,Protein phosphorylation ,ASK1 ,Amino Acid Sequence ,Phosphorylation ,Molecular Biology ,MAPK14 ,Mitogen-Activated Protein Kinase Kinases ,General Immunology and Microbiology ,biology ,MAP kinase kinase kinase ,General Neuroscience ,Cyclin-dependent kinase 2 ,Epithelial Cells ,Cell biology ,Calcium-Calmodulin-Dependent Protein Kinases ,biology.protein ,Mitogen-Activated Protein Kinases ,Protein Kinases ,Anisomycin ,HeLa Cells - Abstract
We have developed a method of general application for identifying putative substrates of protein kinases in cell extracts. Using this procedure, we identified the physiological substrates of several mitogen-activated protein kinase kinases and an authentic substrate of stress-activated protein kinase (SAPK) 2a/p38. A 120 kDa protein was detected in skeletal muscle extracts that was phosphorylated rapidly by SAPK4/p38delta, but poorly by SAPK2/p38, SAPK3/p38gamma, SAPK1/JNK or extracellular signal-regulated kinase 2 (ERK2). It was purified and identified as eukaryotic elongation factor 2 kinase (eEF2K). SAPK4/p38delta phosphorylated eEF2K at Ser359 in vitro, causing its inactivation. eEF2K became phosphorylated at Ser359 and its substrate eEF2 became dephosphorylated (activated) when KB cells were exposed to anisomycin, an agonist that activates all SAPKs, including SAPK4/p38delta. The anisomycin-induced phosphorylation of Ser359 was unaffected by SB 203580, U0126 or rapamycin, and was prevented by overexpression of a catalytically inactive SAPK4/p38delta mutant, suggesting that SAPK4/p38delta may mediate the inhibition of eEF2K by this stress. The phosphorylation of eEF2K at Ser359 was also induced by insulin-like growth factor-1. However, this was blocked by rapamycin, indicating that Ser359 is targeted by at least two signalling pathways.
- Published
- 2001
50. Phosphorylation of serine 230 promotes inducible transcriptional activity of heat shock factor 1
- Author
-
Lea Sistonen, Carina I. Holmberg, Andrey Mikhailov, Jouni O. Rantanen, Carol MacKintosh, John E. Eriksson, Marko J. Kallio, Richard I. Morimoto, Ville Hietakangas, Nick Morrice, Annika Meinander, and Jukka Hellman
- Subjects
Phosphopeptides ,Transcriptional Activation ,Hot Temperature ,Consensus site ,Biology ,environment and public health ,Antibodies ,Article ,General Biochemistry, Genetics and Molecular Biology ,Phosphorylation cascade ,Transactivation ,Heat Shock Transcription Factors ,Ca2+/calmodulin-dependent protein kinase ,Serine ,Tumor Cells, Cultured ,Humans ,Phosphorylation ,Heat shock ,Fluorescent Antibody Technique, Indirect ,HSF1 ,Molecular Biology ,General Immunology and Microbiology ,General Neuroscience ,fungi ,Molecular biology ,Recombinant Proteins ,DNA-Binding Proteins ,Heat shock factor ,enzymes and coenzymes (carbohydrates) ,Calcium-Calmodulin-Dependent Protein Kinases ,Mutagenesis, Site-Directed ,Calcium-Calmodulin-Dependent Protein Kinase Type 2 ,Transcription Factors - Abstract
Heat shock factor 1 (HSF1) is a serine-rich constitutively phosphorylated mediator of the stress response. Upon stress, HSF1 forms DNA-binding trimers, relocalizes to nuclear granules, undergoes inducible phosphorylation and acquires the properties of a transactivator. HSF1 is phosphorylated on multiple sites, but the sites and their function have remained an enigma. Here, we have analyzed sites of endogenous phosphorylation on human HSF1 and developed a phosphopeptide antibody to identify Ser230 as a novel in vivo phosphorylation site. Ser230 is located in the regulatory domain of HSF1, and promotes the magnitude of the inducible transcriptional activity. Ser230 lies within a consensus site for calcium/calmodulin-dependent protein kinase II (CaMKII), and CaMKII overexpression enhances both the level of in vivo Ser230 phosphorylation and transactivation of HSF1. The importance of Ser230 was further established by the S230A HSF1 mutant showing markedly reduced activity relative to wild-type HSF1 when expressed in hsf1(-/-) cells. Our study provides the first evidence that phosphorylation is essential for the transcriptional activity of HSF1, and hence for induction of the heat shock response.
- Published
- 2001
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.