Volume 63, Issue 3, Pages (August 2016)

Slides:



Advertisements
Similar presentations
Volume 41, Issue 6, Pages (March 2011)
Advertisements

Volume 22, Issue 4, Pages e4 (October 2017)
Volume 9, Issue 5, Pages (November 1998)
Volume 41, Issue 2, Pages (January 2011)
Takashi Tanaka, Michelle A. Soriano, Michael J. Grusby  Immunity 
Volume 33, Issue 2, Pages (January 2009)
Volume 45, Issue 5, Pages (November 2016)
Shitao Li, Lingyan Wang, Michael A. Berman, Ye Zhang, Martin E. Dorf 
Volume 44, Issue 4, Pages (November 2011)
Yu-Hsin Chiu, Jennifer Y. Lee, Lewis C. Cantley  Molecular Cell 
Volume 15, Issue 6, Pages (June 2012)
Volume 11, Issue 6, Pages (June 2012)
Volume 22, Issue 5, Pages (May 2012)
Volume 30, Issue 3, Pages (March 2009)
Monica C. Rodrigo-Brenni, Erik Gutierrez, Ramanujan S. Hegde 
Volume 26, Issue 2, Pages (February 2007)
Volume 35, Issue 6, Pages (December 2011)
Volume 20, Issue 3, Pages (September 2016)
DAI/ZBP1/DLM-1 Complexes with RIP3 to Mediate Virus-Induced Programmed Necrosis that Is Targeted by Murine Cytomegalovirus vIRA  Jason W. Upton, William J.
Yongli Bai, Chun Yang, Kathrin Hu, Chris Elly, Yun-Cai Liu 
Volume 45, Issue 5, Pages (March 2012)
T helper17 Cells Are Sufficient But Not Necessary to Induce Acute Graft-Versus-Host Disease  Cristina Iclozan, Yu Yu, Chen Liu, Yaming Liang, Tangsheng.
Volume 46, Issue 4, Pages (April 2017)
MUC1 Oncoprotein Stabilizes and Activates Estrogen Receptor α
Volume 16, Issue 1, Pages (January 2006)
Anti-Inflammatory Activity of Sertaconazole Nitrate Is Mediated via Activation of a p38– COX-2–PGE2 Pathway  Runa Sur, Jeffrey M. Babad, Michelle Garay,
Volume 125, Issue 4, Pages (May 2006)
Xiaolong Wei, Hai Xu, Donald Kufe  Cancer Cell 
Volume 20, Issue 4, Pages (April 2004)
Volume 22, Issue 4, Pages (April 2005)
Volume 15, Issue 6, Pages (June 2012)
MUC1 Oncoprotein Stabilizes and Activates Estrogen Receptor α
SGK3 Mediates INPP4B-Dependent PI3K Signaling in Breast Cancer
Active Caspase-1 Is a Regulator of Unconventional Protein Secretion
Volume 13, Issue 1, Pages (January 2008)
TNF-Induced Activation of the Nox1 NADPH Oxidase and Its Role in the Induction of Necrotic Cell Death  You-Sun Kim, Michael J. Morgan, Swati Choksi, Zheng-gang.
Volume 66, Issue 4, Pages e5 (May 2017)
Volume 33, Issue 2, Pages (August 2010)
HDAC5, a Key Component in Temporal Regulation of p53-Mediated Transactivation in Response to Genotoxic Stress  Nirmalya Sen, Rajni Kumari, Manika Indrajit.
Volume 25, Issue 5, Pages (November 2006)
Septins Regulate Actin Organization and Cell-Cycle Arrest through Nuclear Accumulation of NCK Mediated by SOCS7  Brandon E. Kremer, Laura A. Adang, Ian.
The Actin-Bundling Protein Palladin Is an Akt1-Specific Substrate that Regulates Breast Cancer Cell Migration  Y. Rebecca Chin, Alex Toker  Molecular.
Volume 15, Issue 4, Pages (August 2004)
Volume 38, Issue 3, Pages (March 2013)
Volume 17, Issue 6, Pages (June 2015)
Volume 21, Issue 6, Pages (November 2017)
E3 Ubiquitin Ligase VHL Regulates Hypoxia-Inducible Factor-1α to Maintain Regulatory T Cell Stability and Suppressive Capacity  Jee H. Lee, Chris Elly,
Volume 25, Issue 5, Pages (March 2007)
Mst1 Is an Interacting Protein that Mediates PHLPPs' Induced Apoptosis
Volume 30, Issue 6, Pages (June 2009)
Volume 43, Issue 4, Pages (October 2015)
Volume 34, Issue 5, Pages (May 2011)
Volume 32, Issue 2, Pages (February 2010)
Volume 16, Issue 1, Pages (January 2006)
Volume 41, Issue 2, Pages (August 2014)
USP15 Negatively Regulates Nrf2 through Deubiquitination of Keap1
NF-κB Is Required for UV-Induced JNK Activation via Induction of PKCδ
In Vitro Analysis of Huntingtin-Mediated Transcriptional Repression Reveals Multiple Transcription Factor Targets  Weiguo Zhai, Hyunkyung Jeong, Libin.
Volume 16, Issue 16, Pages (August 2006)
Volume 5, Issue 4, Pages (April 2007)
E3-Independent Monoubiquitination of Ubiquitin-Binding Proteins
Suman Paul, Anuj K. Kashyap, Wei Jia, You-Wen He, Brian C. Schaefer 
Volume 45, Issue 5, Pages (November 2016)
Volume 23, Issue 2, Pages (August 2005)
A Direct HDAC4-MAP Kinase Crosstalk Activates Muscle Atrophy Program
Volume 57, Issue 2, Pages (January 2015)
Volume 65, Issue 5, Pages e4 (March 2017)
Volume 37, Issue 2, Pages (August 2012)
Volume 10, Issue 5, Pages (February 2015)
Volume 50, Issue 5, Pages (June 2013)
Presentation transcript:

Volume 63, Issue 3, Pages 457-469 (August 2016) Identification of PGAM5 as a Mammalian Protein Histidine Phosphatase that Plays a Central Role to Negatively Regulate CD4+ T Cells  Saswati Panda, Shekhar Srivastava, Zhai Li, Martin Vaeth, Stephen R. Fuhs, Tony Hunter, Edward Y. Skolnik  Molecular Cell  Volume 63, Issue 3, Pages 457-469 (August 2016) DOI: 10.1016/j.molcel.2016.06.021 Copyright © 2016 Elsevier Inc. Terms and Conditions

Molecular Cell 2016 63, 457-469DOI: (10.1016/j.molcel.2016.06.021) Copyright © 2016 Elsevier Inc. Terms and Conditions

Figure 1 PGAM5 Specifically Interacts with NDPK-B and Functions as a Histidine Phosphatase to Dephosphorylate H118 on NDPK-B (A) Whole-cell lysates from HEK293T cells overexpressing FLAG-NDPK-B and myc-PGAM5 (WT or H105A mutant) were immunoprecipitated (IP) with anti-FLAG or anti-myc antibodies followed by immunoblotting (IB) with anti-myc or anti-FLAG antibodies as indicated. (B) CoIP of endogenous NDPK-B and PGAM5 in HEK293T cells. Lysates from control or PGAM5 shRNA-transfected cells were subjected to IP with isotype control antibody or NDPK-B or PGAM5 antibodies, and blots were probed as indicated. The association was specific as demonstrated by the failure of either NDPK-B or PGAM5 to be IP with isotype control antibodies and the failure of anti-PGAM5 antibodies to coIP NDPK-B in PGAM5 shRNA-transfected cells. (C) FLAG-NDPK-A, NDPK-B, or NDPK-C was transfected into HEK293T cells together with myc-PGAM5WT, and lysates were subjected to anti-FLAG IP followed by IB with anti-myc or anti-FLAG antibodies as shown. (D) Purified GST-tagged NDPK-BWT was auto-phosphorylated with GTP and then incubated with His-tagged PGAM5WT or PGAM5 mutant (PGAM5H105A) for various times as indicated. Purity of His-tagged PGAM5 proteins was assessed by Coomassie blue staining (Figure S2A). Samples were resolved by SDS-PAGE under basic conditions and immunoblotted with anti-1-pHis, 3-pHis, NDPK-B, and PGAM5 antibodies as indicated. (E) FLAG-NDPK-B (WT or H118N) was transfected into 293T cells along with myc-PGAM5WT or PGAM5H105A. Lysates were IP with anti-FLAG antibodies and IB with anti-1-pHis and anti-NDPK-B antibodies. Relative levels of overexpressed PGAM5 and NDPK-B were 3- to 5-fold higher compared to the endogenous protein levels (Figure S2B). (F) 293T cells were transfected with FLAG-NDPK-A or NDPK-B along with myc-PGAM5 (WT or H105A mutant). Lysates were subjected to anti-FLAG IP and IB with anti-FLAG and anti-1-pHis antibodies (to detect NDPK phosphorylation). Lysates were probed with anti-myc and anti β-actin antibodies as indicated. Data are representative of three independent experiments. See also Figures S1, S2, and S7. Molecular Cell 2016 63, 457-469DOI: (10.1016/j.molcel.2016.06.021) Copyright © 2016 Elsevier Inc. Terms and Conditions

Figure 2 PGAM5 Negatively Regulates KCa3.1 Activity in Jurkat T Cells (A) Jurkat T cells overexpressing GFP-KCa3.1 (Jurkat-KCa3.1 cells) were transfected with PGAM5 or control siRNA. Lysates were IP with anti-NDPK-B or anti-GFP (KCa3.1) antibodies and IB with antibodies to 1-pHis, 3-pHis, NDPK-B, and GFP antibodies as indicated. Half of the IPs also were heated at 80°C for 2 min prior to loading to ensure specificity of detection of histidine phosphorylation (histidine phosphorylation is heat sensitive). Lysates were probed with antibodies to GFP (KCa3.1), NDPK-B, PGAM5 (to demonstrate knockdown of PGAM5), and β-actin (as a loading control). (B) KCa3.1 channel activity (TRAM-34 sensitive) was determined by whole-cell patch clamp on Jurkat-KCa3.1 T cells transfected with (i) control or (ii) PGAM5 siRNA. Shown are current/voltage (I/V) plots of the cells, as previously described (Srivastava et al., 2006a). TRAM-34 is a specific inhibitor of KCa3.1 channel activity. (C) Bar graph summary shows TRAM-34-sensitive current in cells transfected with control or PGAM5 siRNA and PGAM5-knockdown cells rescued with siRNA-resistant PGAM5 (WT or H105A mutant), plotted at +60 mV (n = 15–20 cells each). (D) Calcium flux was performed in cells loaded with Fura-2 AM (5 μM), attached to a poly-L-lysine-coated coverslip and re-stimulated with anti-CD3. Ca2+ imaging was done with an IX81 epifluorescence microscope (Olympus), and data were analyzed using OpenLab imaging software (Improvision) (n = 80–100 cells for each series), as previously described (Srivastava et al., 2009). (E) ELISA to quantify IL-2 in the supernatants of control or PGAM5 siRNA-transfected cells at 48 hr post-transfection is shown. Data are representative of three independent experiments. Data are shown as mean ± SEM. Statistical significance was calculated using Student’s t test (∗p < 0.05 and ∗∗p < 0.01; n.s. (p > 0.05), not significant). See also Figure S3. Molecular Cell 2016 63, 457-469DOI: (10.1016/j.molcel.2016.06.021) Copyright © 2016 Elsevier Inc. Terms and Conditions

Figure 3 PGAM5 Does Not Directly Dephosphorylate or Inactivate KCa3.1 (A) GFP-tagged KCa3.1 was IP with anti-GFP antibody from 293-KCa3.1 GFP cells and incubated with purified His-tagged PGAM5 (WT or H105A mutant) or His-tagged PHPT-1 for various time points as indicated. Samples were resolved by SDS-PAGE under basic conditions (pH 8.8) and IB with 3-pHis and GFP antibodies as indicated. (B) (i) Inside-out (I/O) patches were isolated from 293-KCa3.1 cells. Single-channel activity was then recorded in I/O patches that were first incubated with GST-NDPK-B in the presence of 300 nM Ca2+ and GTP. This was followed by the addition of His-tagged PGAM5 and His-tagged PHPT-1 to the same patch as indicated in the trace. (ii) Effect of NDPK-B, PGAM5, and PHPT-1 on the open-channel probability, NPo, is shown. Bar graph represents KCa3.1 NPo as described in (i). All recordings were at +100 mV. Data are representative of three independent experiments. Data are shown as mean ± SEM. Statistical significance was calculated using Student’s t test (∗p < 0.05; n.s. (p > 0.05), not significant. Molecular Cell 2016 63, 457-469DOI: (10.1016/j.molcel.2016.06.021) Copyright © 2016 Elsevier Inc. Terms and Conditions

Figure 4 PGAM5 Negatively Regulates Primary Human CD4+ T Cells (A–E) Primary human naive CD4+ T cells were transfected with PGAM5 or control siRNA, rested overnight, and then activated with anti-CD3 and anti-CD28 antibodies for 48 hr to generate Th0 CD4+ T cells. (A) Lysates were IP with anti-NDPK-B, and NDPK-B phosphorylation was assessed by IB with anti-1-pHis antibodies. Lysates also were probed with anti-PGAM5 and anti-β-actin antibodies to verify PGAM5 knockdown. (B) Whole-cell patch clamp was performed on cells to determine KCa3.1 and Kv1.3 activity. Shown are I/V plots of the cells transfected with (i) control or (ii) PGAM5 siRNA. (iii) Scatterplot of TRAM-34-sensitive current plotted at +60 mV (n = 15–20 cells each). TRAM-34 and ShK are specific inhibitors of KCa3.1 and Kv1.3 channel activity, respectively (Srivastava et al., 2006a). (C) Th0 cells were rested overnight, loaded with Fura-2 AM (5 μM), attached to a poly-L-lysine-coated coverslip, and re-stimulated with anti-CD3. Ca2+ imaging was done with an IX81 epifluorescence microscope (Olympus), and data were analyzed using OpenLab imaging software (Improvision) (n = 80–100 cells for each series), as previously described (Srivastava et al., 2009). (D) Quantification of soluble IL-2 secreted by Th0 cells rested overnight and re-stimulated with anti-CD3/CD28 antibodies for 6 hr is shown. (E) Representative intracellular flow cytometry detecting cytokine expression in cells that were rested overnight and re-stimulated with phorbol 12-myristate 13-acetate (PMA)/ionomycin for 4 hr (left panel). Shown are percentage positive (middle panel) and mean fluorescence intensity (MFI) (right panel) of cytokine-producing cells. Data are representative of three independent experiments. Data are shown as mean ± SEM. Statistical significance was calculated using Student’s t test (∗p < 0.05 and ∗∗p < 0.01; n.s. (p > 0.05), not significant. See also Figure S4. Molecular Cell 2016 63, 457-469DOI: (10.1016/j.molcel.2016.06.021) Copyright © 2016 Elsevier Inc. Terms and Conditions

Figure 5 PGAM5 Counteracts Human CD4+ T Cell Activation through NDPK-B Dephosphorylation (A–C) Primary human naive CD4+ T cells were transfected with a control siRNA, siRNA pool to PGAM5, NDPK-B, or both, rested overnight, and then activated with anti-CD3 and anti-CD28 antibodies for 48 hr (to generate Th0 CD4+ T cells). (A) NDPK-B was IP from cell lysates and IB with antibodies to 1-pHis and NDPK-B to assess NDPK-B 1-pHis phosphorylation. Lysates were IB with anti-NDPK-B and anti-PGAM5 to demonstrate knockdown of each protein and anti-β-actin was used as a loading control. (B) Whole-cell patch clamp of cells as described in Figure 4B. Shown is a bar graph summary of TRAM-34-sensitive and Shk-sensitive currents plotted at +60 mV (n = 15–20 cells each). (C) Ca2+ flux was determined as shown in Figure 4C (n = 80–100 cells for each series). Data are representative of three independent experiments. Data are shown as mean ± SEM. Statistical significance was calculated using Student’s t test (∗p < 0.05 and ∗∗p < 0.01; n.s. (p > 0.05), not significant. Molecular Cell 2016 63, 457-469DOI: (10.1016/j.molcel.2016.06.021) Copyright © 2016 Elsevier Inc. Terms and Conditions

Figure 6 PGAM5 Negatively Regulates Activation of Mouse CD4+ T Cells (A–E) Naive CD4+ T cells were isolated from spleens of WT or Pgam5−/− mice and activated with anti-CD3 and anti-CD28 antibodies for 48 hr to generate Th0 cells. (A) Naive or Th0 WT or Pgam5−/− cell lysates were subjected to IP with anti-NDPK-B and anti-PGAM5 antibodies and IB as indicated. The failure of anti-PGAM5 antibodies to coIP NDPK-B from Pgam5−/− naive and Th0 cells demonstrates the interaction is specific, as PGAM5 needs to be present for anti-PGAM5 antibodies to coIP NDPK-B. (B) NDPK-B and KCa3.1 were IP from WT and Pgam5−/− naive or Th0 cell lysates, and then probed with antibodies to 1-pHis and NDPK-B for the NDPK-B IP and 3-pHis and KCa3.1 for the KCa3.1 IP. KCa3.1 is expressed at low levels in naive cells and is induced following activation with anti-CD3/CD28 antibodies (Th0 cells). Th0 cell lysates were blotted with NDPK-B, PGAM5, and β-actin as a loading control. (C) Whole-cell patch clamp of Th0 cells to determine KCa3.1 activity as described in Figure 4B. Shown are I/V plots of (i) WT and (ii) Pgam5−/− cells. (iii) Scatterplot shows TRAM-34- and Shk-sensitive currents (n = 15–20 cells each). (D) Th0 cells were rested overnight and then re-stimulated with anti-CD3 for measuring calcium flux as described in Figure 4C (n = 80–100 cells for each series). (E) Representative intracellular flow cytometry detecting cytokine expression of WT or Pgam5−/− CD4+ T cells. Th0 cells were rested overnight and re-stimulated with PMA/ionomycin for 4 hr (left panel). Shown are percentage positive (middle panel) and MFI (right panel) of cytokine-producing cells. Data are representative of three independent experiments. Data are shown as mean ± SEM (n = 3 mice per group per experiment). Statistical significance was calculated using Student’s t test (∗p < 0.05 and ∗∗p < 0.01; n.s. (p > 0.05), not significant. See also Figure S5. Molecular Cell 2016 63, 457-469DOI: (10.1016/j.molcel.2016.06.021) Copyright © 2016 Elsevier Inc. Terms and Conditions

Figure 7 Pgam5−/− T Cells Cause Augmented Pathogenicity after Allogeneic Transplantation (A–E) Host BALB/c mice (H-2d) were lethally irradiated and transplanted with either 1.5 × 106 allogeneic WT or Pgam5−/− T cells isolated from spleens of donor C57BL/6 (H-2b), along with 5 × 106 bone marrow (BM) cells. BM control group received only BM cells. (A) Body weight changes of host mice groups post-allogeneic-hematopoietic cell transfer (HCT) are shown (n = 15 mice per group). (B) Clinical GvHD scoring (score 0–8) of mice groups post-HCT is shown (n = 15 mice per group). (C) Serum IFNγ and TNF-α analysis at day 6 post-HCT using ELISA. Data are compiled from two independent experiments (n = 5 mice per group per experiment). (D) Survival of mice groups post-HCT is shown (n = 15 mice per group). (E) CD4+ T cells were purified from spleens of host mice at day 6 post-HCT, and calcium flux was performed as described in Figure 4C (n = 80–100 cells for each series). Data shown are representative of two independent experiments. Statistical significance was calculated using Student’s t test (Figure 7C) (∗∗∗p < 0.001) and log-rank (Mantel-Cox) test (Figure 7D) (∗∗∗∗p < 0.0001). See also Figure S6. Molecular Cell 2016 63, 457-469DOI: (10.1016/j.molcel.2016.06.021) Copyright © 2016 Elsevier Inc. Terms and Conditions