A novel TNFR1-triggered apoptosis pathway mediated by class IA PI3Ks in neutrophils by Barbara Geering, Ursina Gurzeler, Elena Federzoni, Thomas Kaufmann,

Slides:



Advertisements
Similar presentations
Volume 54, Issue 4, Pages (May 2014)
Advertisements

Induction of Apoptosis by Ethanol Extracts of Ganoderma lucidum in Human Gastric Carcinoma Cells  Kyung-Jun Jang, Min-Ho Han, Byung-Hoon Lee, Byung-Woo.
Phosphatidylinositol 3-kinase and p38 mitogen-activated protein kinase regulate induction of Mcl-1 and survival in glucocorticoid-treated human neutrophils 
by Yunping Lin, Lauren Brown, David W. Hedley, Dwayne L
Volume 125, Issue 3, Pages (September 2003)
Volume 129, Issue 2, Pages (August 2005)
Volume 129, Issue 1, Pages (July 2005)
G-CSF improves murine G6PC3-deficient neutrophil function by modulating apoptosis and energy homeostasis by Hyun Sik Jun, Young Mok Lee, Ki Duk Song, Brian.
The CXC-chemokine platelet factor 4 promotes monocyte survival and induces monocyte differentiation into macrophages by Barbara Scheuerer, Martin Ernst,
Enhancement of intracellular signaling associated with hematopoietic progenitor cell survival in response to SDF-1/CXCL12 in synergy with other cytokines.
Dual-targeting immunotherapy of lymphoma: potent cytotoxicity of anti-CD20/CD74 bispecific antibodies in mantle cell and other lymphomas by Pankaj Gupta,
by Fawzi Aoudjit, and Kristiina Vuori
Constitutively activated phosphatidylinositol-3 kinase (PI-3K) is involved in the defect of apoptosis in B-CLL: association with protein kinase Cδ by Ingo.
Differential effects of tumor necrosis factor-α and interleukin-1β on cell death in human articular chondrocytes  B. Caramés, Ph.D., M.J. López-Armada,
Lipopolysaccharide Activates Caspase-1 (Interleukin-1–Converting Enzyme) in Cultured Monocytic and Endothelial Cells by Ralf R. Schumann, Claus Belka,
by Kumudha Balakrishnan, William G. Wierda, Michael J
by Fan Dong, and Andrew C. Larner
Phosphatidylinositol 3-kinase and p38 mitogen-activated protein kinase regulate induction of Mcl-1 and survival in glucocorticoid-treated human neutrophils 
by Christina K. Ullrich, Jerome E. Groopman, and Ramesh K. Ganju
Proviral integration site for Moloney murine leukemia virus 1, but not phosphatidylinositol-3 kinase, is essential in the antiapoptotic signaling cascade.
Activation of the PKB/AKT Pathway by ICAM-2
Caspases Mediate Tumor Necrosis Factor-–Induced Neutrophil Apoptosis and Downregulation of Reactive Oxygen Production by Kouhei Yamashita, Atsushi Takahashi,
Activation of ATF4 mediates unwanted Mcl-1 accumulation by proteasome inhibition by Jinsong Hu, Nana Dang, Eline Menu, Elke De Bryune, Dehui Xu, Ben Van.
Small-molecule inhibitor QLT-0267 suppresses ILK activity and inhibits its downstream signaling. Small-molecule inhibitor QLT-0267 suppresses ILK activity.
Phosphoinositide 3-kinase inhibitors protect mouse kidney cells from cyclosporine- induced cell death  E. Sarró, O. Tornavaca, M. Plana, A. Meseguer, E.
by Thomas T. Murooka, Ramtin Rahbar, Leonidas C
Sustained signaling through the B-cell receptor induces Mcl-1 and promotes survival of chronic lymphocytic leukemia B cells by Aleksandar Petlickovski,
Volume 70, Issue 6, Pages (September 2006)
by Mi-Ae Kang, Su-Young Yun, and Jonghwa Won
H.T. Lee, M. Kim, M. Jan, R.B. Penn, C.W. Emala  Kidney International 
Cathepsin-B-dependent apoptosis triggered by antithymocyte globulins: a novel mechanism of T-cell depletion by Marie-Cécile Michallet, Frederic Saltel,
The PI3K/mTOR inhibitor PF induces apoptosis and inhibits microenvironmental signaling in CLL and the Eµ-TCL1 mouse model by Matthew D. Blunt,
by Juan M. Cárcamo, Oriana Bórquez-Ojeda, and David W. Golde
by Asim Khwaja, and Louise Tatton
Cell-surface CD74 initiates a signaling cascade leading to cell proliferation and survival by Diana Starlets, Yael Gore, Inbal Binsky, Michal Haran, Nurit.
Phosphatidylinositol 3-Kinase/Akt-Dependent and -Independent Protection Against Apoptosis in Normal Human Melanocytes  Masahiro Oka, Akiko Kageyama, Mizuho.
AKT Delays the Early-Activated Apoptotic Pathway in UVB-Irradiated Keratinocytes Via BAD Translocation  Sofie Claerhout, David Decraene, An Van Laethem,
Rapid ubiquitination of Syk following GPVI activation in platelets
Volume 141, Issue 2, Pages (August 2011)
by Xingwei Sui, Sanford B. Krantz, Min You, and Zhizhuang Zhao
LPS induces CD40 gene expression through the activation of NF-κB and STAT-1α in macrophages and microglia by Hongwei Qin, Cynthia A. Wilson, Sun Jung Lee,
Small-molecule XIAP inhibitors derepress downstream effector caspases and induce apoptosis of acute myeloid leukemia cells by Bing Z. Carter, Marcela Gronda,
Regulation of Akt-dependent cell survival by Syk and Rac
Critical role for PI 3-kinase in the control of erythropoietin-induced erythroid progenitor proliferation by Didier Bouscary, Frédéric Pene, Yann-Erick.
by Madelon Bracke, Evert Nijhuis, Jan-Willem J. Lammers, Paul J
Volume 122, Issue 5, Pages (May 2002)
Stimulation of the B-cell receptor activates the JAK2/STAT3 signaling pathway in chronic lymphocytic leukemia cells by Uri Rozovski, Ji Yuan Wu, David.
Volume 136, Issue 4, Pages e3 (April 2009)
Volume 56, Issue 4, Pages (October 1999)
by Reuben Kapur, Ryan Cooper, Lei Zhang, and David A. Williams
Akio Horiguchi, Mototsugu Oya, Ken Marumo, Masaru Murai 
Involvement of Fas (APO-1/CD-95) during Photodynamic-Therapy-Mediated Apoptosis in Human Epidermoid Carcinoma A431 Cells  Nihal Ahmad, Sanjay Gupta, Denise.
Mechanisms of cross hyporesponsiveness to toll-like receptor bacterial ligands in intestinal epithelial cells  Jan-Michel Otte, Elke Cario, Daniel K.
Volume 125, Issue 1, Pages (July 2003)
Volume 9, Issue 3, Pages (November 2014)
Upregulation of Tenascin-C Expression by IL-13 in Human Dermal Fibroblasts via the Phosphoinositide 3-kinase/Akt and the Protein Kinase C Signaling Pathways 
Volume 11, Issue 6, Pages (June 2003)
Honglin Li, Hong Zhu, Chi-jie Xu, Junying Yuan  Cell 
Differential Regulation of Cyclooxygenase-2 Expression by Phytosphingosine Derivatives, NAPS and TAPS, and its Role in the NAPS or TAPS-Mediated Apoptosis 
Volume 122, Issue 1, Pages (January 2002)
Volume 8, Issue 4, Pages (October 2005)
Tumor necrosis factor-α and lipopolysaccharide induce apoptotic cell death in bovine glomerular endothelial cells  Udo K. Meßmer, Verena A. Briner, Josef.
Volume 61, Issue 2, Pages (February 2002)
Volume 70, Issue 5, Pages (September 2006)
Nan-Lin Wu, Te-An Lee, Te-Lung Tsai, Wan-Wan Lin 
Adhesion-induced eosinophil cytolysis requires the receptor-interacting protein kinase 3 (RIPK3)–mixed lineage kinase-like (MLKL) signaling pathway, which.
Volume 61, Issue 2, Pages (February 2002)
The Akt/Mcl-1 pathway plays a prominent role in mediating antiapoptotic signals downstream of the B-cell receptor in chronic lymphocytic leukemia B cells.
Lamellarin D induces cell death through a Fas-independent pathway.
by Dana S. Levy, Jason A. Kahana, and Rakesh Kumar
Presentation transcript:

A novel TNFR1-triggered apoptosis pathway mediated by class IA PI3Ks in neutrophils by Barbara Geering, Ursina Gurzeler, Elena Federzoni, Thomas Kaufmann, and Hans-Uwe Simon Blood Volume 117(22):5953-5962 June 2, 2011 ©2011 by American Society of Hematology

TNF-α induces neutrophil apoptosis independently of Bid. TNF-α induces neutrophil apoptosis independently of Bid. (A) TNF-α stimulation results in neutrophil cell death. Primary human neutrophils were stimulated with TNF-α (50 ng/mL) or anti-FAS antibody (1 μg/mL). Twenty-four hours after neutrophil isolation, viability was assessed by ethidium bromide staining and flow cytometry (n > 10) (left panel). Eight hours after neutrophil isolation, neutrophils were harvested on glass plates, fixed, and stained. Neutrophils with apoptotic features are marked with arrows. A representative section in 1 of 4 independent experiments is shown (right panel). (B) TNF-α–induced murine neutrophil death is independent of Bid. After isolation of neutrophils from wild-type or bid−/− mice, cells were left untreated or stimulated with TNF-α (50 ng/mL) or FAS ligand (100 ng/mL). After 24 hours of culture, neutrophil apoptosis was assessed by annexin V–propidium iodide staining and flow cytometry (n = 5). (C) Cytochrome c is not released in TNF-α–stimulated neutrophils. Primary human neutrophils were stimulated with TNF-α or anti-FAS antibody for 4 hours. Cytochrome (Cyt) c release from mitochondria was analyzed by flow cytometry. Shown are a quantification of cytochrome c release (n = 3) (left panel) and representative flow cytometry diagrams (right panel). Barbara Geering et al. Blood 2011;117:5953-5962 ©2011 by American Society of Hematology

TNF-α–induced neutrophil apoptosis is dependent on effector caspase activity. TNF-α–induced neutrophil apoptosis is dependent on effector caspase activity. (A) Caspase inhibition before TNF-α stimulation results in neutrophil survival. Primary human neutrophils were pretreated with vehicle (Medium) or 10 or 50μM pan-caspase inhibitor VAD in control, TNF-α-treated, and anti-FAS antibody–treated neutrophils. Viability was assessed by ethidium bromide staining and flow cytometry after 24 hours of stimulation (n = 3 for 50μM VAD; n = 7 for 10μM VAD). (B) Caspase-8 is not activated after TNF-α stimulation. Caspase-8 activity was assessed by immunoblot analysis of BID processing. Neutrophils were harvested immediately (Fresh) or left untreated (Control) and stimulated with TNF-α (20 ng/mL) or anti-FAS antibody (1 μg/mL) for 4 hours. A representative immunoblot is shown and BID protein expression levels were quantified relative to the fresh sample (n = 3). (C) Effector caspases are activated after TNF-α stimulation. Total effector caspase activity was assessed by immunoblot analysis of lamin B processing. Neutrophils were treated as in panel B. A representative immunoblot is shown. Lamin B protein expression levels were quantified relative to the 67-kDa lamin B band in the fresh sample (n = 2). Barbara Geering et al. Blood 2011;117:5953-5962 ©2011 by American Society of Hematology

TNF-α–induced neutrophil apoptosis is dependent on MAPK p38 and class IA PI3K activity. TNF-α–induced neutrophil apoptosis is dependent on MAPK p38 and class IA PI3K activity. (A) Inhibition of p38 and PI3K before TNF-α stimulation results in neutrophil survival. Before TNF-α (50 ng/mL) or anti-FAS antibody (1 μg/mL) stimulation, primary human neutrophils were incubated for 30-60 minutes with vehicle (Medium) or with small-molecule inhibitors against PI3K (100nM wortmannin), p38 (1μM SB203580), MEK (50μM PD98059), SAPK/JNK (10μM SP600125), or JAK/STAT (25μM AG490). Viability was assessed by ethidium bromide staining and flow cytometry after 24 hours (n ≥ 3). (B) Class IA PI3Ks are death molecules after TNF-α stimulation. Neutrophils were incubated with vehicle (Control, Medium) or different PI3K inhibitors before TNF-α (50 ng/mL) stimulation. Viability was assessed by ethidium bromide staining and flow cytometry after 24 hours. The following inhibitors were used: the broad-spectrum PI3K inhibitor wortmannin (100nM), the class IA PI3K-selective inhibitor PI103 (100nM), the p110β isoform-selective inhibitor TGX221 (100nM), the p110δ isoform-selective inhibitor IC87114 (1μM), and the p110γ isoform-selective inhibitor AS604850 (1 and 10μM) (n ≥ 4). (C) Activation of p38 and PI3Ks is independent of caspase activity. Neutrophils were pretreated with or without VAD for 60 minutes and subsequently stimulated for 15 minutes with 50 ng/mL of TNF-α or 1 μg/mL of anti-FAS antibody. Cell lysates were analyzed by immunoblotting for phosphorylated Ser473 AKT (an indirect readout for PI3K activity) or phosphorylated Thr180/Tyr182 p38. p38, AKT, and GAPDH protein levels were analyzed as loading controls. A representative immunoblot is shown, and protein expression levels were quantified relative to the control condition (n = 3). Barbara Geering et al. Blood 2011;117:5953-5962 ©2011 by American Society of Hematology

PI3Ks are activated by p38 downstream of TNFR1. PI3Ks are activated by p38 downstream of TNFR1. (A) p38 and PI3Ks are activated downstream of TNFR1. Neutrophils were kept untreated (Control, Medium) or incubated with 10 μg/mL of anti-TNFR1 or anti-TNFR2 antibodies for 90 minutes. Cells were subsequently stimulated with 50 ng/mL of TNF-α for 15 minutes and cell lysates were analyzed as in Figure 3C. A representative immunoblot is shown, and protein expression levels were quantified relative to the control condition (n = 3). (B) Neither class IA PI3Ks nor p38 is associated with activated TNFR1. Human neutrophils were stimulated with 20 ng/mL of TNF-α for 5 minutes. The lysates were incubated with anti-TNFR1 antibody (H398) or IgG2a control antibody. Immunoblots were probed for p38, the PI3K class IA regulatory subunits p85α/p85β (p85), TRADD, and GAPDH (n = 3). IP indicates immunoprecipitation. (C) p38 is upstream of PI3K activation. Neutrophils were treated for 90 minutes with vehicle (Control, Medium), with the PI3K inhibitors wortmannin (100nM) and PI103 (100nM), or with the p38 inhibitors SB203580 (1μM) and PD169316 (10μM) before TNF-α (50 ng/mL) stimulation. Immunoblots were probed for phosphorylated Thr308 and Ser473 AKT, phosphorylated Thr180 and Tyr182 p38, phosphorylated Thr334 MK2, and GAPDH. A representative immunoblot is shown. Protein expression levels were quantified relative to the control condition (n ≥ 3). (D) Phosphorylation of p38 precedes phosphorylation of AKT. Neutrophils were stimulated for the indicated times with 50 ng/mL of TNF-α. Cell lysates were analyzed by immunoblotting for phosphorylated Ser473 AKT or phosphorylated Thr180/Tyr182 p38. GAPDH protein levels were analyzed as loading controls. A representative immunoblot is shown. Protein expression levels were quantified relative to the control condition (n = 3). Barbara Geering et al. Blood 2011;117:5953-5962 ©2011 by American Society of Hematology

p38 and class IA PI3K mediate neutrophil apoptosis by controlling ROS generation. p38 and class IA PI3K mediate neutrophil apoptosis by controlling ROS generation. (A) Inhibition of p38 or PI3Ks before TNF-α stimulation abolishes ROS generation. Time course of ROS generation after TNF-α and FAS antibody stimulation. Neutrophils were pretreated with vehicle or 100nM wortmannin, 10μM SB203580, or 20μM of the NADPH oxidase inhibitor DPI before TNF-α stimulation (50 ng/mL). Cells were also treated with anti-FAS antibody (n > 5). (B) Class IA PI3Ks control ROS generation after TNF-α stimulation. Neutrophils were treated with vehicle (Control, Medium), diverse class IA PI3K inhibitors (for concentrations, see Figure 3B), or 10μM of the pan-caspase inhibitor VAD before TNF-α stimulation (50 ng/mL) (n > 5, except pretreatment with AS604850, where n = 2). (C) Inhibition of PI3Ks does not affect the viability of neutrophils derived from CGD patients. Neutrophils from 3 different CGD patients (who lack a functional NADPH oxidase) were analyzed for ROS generation (left panel) and for viability (right panel; ethidium bromide staining and flow cytometry) after PI3K or NADPH oxidase inhibition and TNF-α stimulation. Barbara Geering et al. Blood 2011;117:5953-5962 ©2011 by American Society of Hematology

Essential role of ROS for caspase-3 activity after TNF-α stimulation. Essential role of ROS for caspase-3 activity after TNF-α stimulation. (A) Inhibition of p38 or PI3Ks does not affect caspase-8 or caspase-3 cleavage. Primary human neutrophils were incubated in the presence or absence of the p38 inhibitor SB203580 (1μM) or the PI3K inhibitor wortmannin (100nM) for 1 hour, and then stimulated with TNF-α (50 ng/mL) or anti-FAS antibody. Cells were harvested after 4 hours for analysis of caspase-8 processing (left panel) or 8 hours for analysis of caspase-3 processing (right panel) of culture. A representative immunoblot is shown, and protein expression levels of the active fragment p18 and p17, respectively, were quantified relative to the fresh sample (n = 4). (B) Caspase-3, but not caspase-8, activity is controlled by ROS. Caspase-8 (IETDase) and caspase-3 (DEVDase) activities were assessed by a colorimetric or fluorometric in vitro caspase activity assay. Neutrophils were harvested immediately after isolation (Fresh), left untreated (Control), or incubated with 20μM DPI, 100nM wortmannin, or 1μM SB203580 before stimulation with TNF-α (50 ng/mL). Neutrophils were also stimulated with 1 μg/mL of anti-FAS antibody. Cells were harvested and lysed at 4 hours for analysis of caspase-8 activity or at 8 hours for analysis of caspase-3 activity. Caspase-8 and caspase-3 activities are shown relative to the caspase activity detected in fresh neutrophil lysates (set as 1) (n ≥ 3). Barbara Geering et al. Blood 2011;117:5953-5962 ©2011 by American Society of Hematology

Working model of TNF-α- and FAS ligand–induced neutrophil apoptosis. Working model of TNF-α- and FAS ligand–induced neutrophil apoptosis. Left panel shows that TNFR1 ligation results in p38 activation. p38 further activates the PI3K signaling pathway and ROS generation. ROS increase caspase-3 activity, leading to apoptosis. Right panel shows that ligation of FAS results in caspase-8 processing, which further activates caspase-3 and BID. BID cleavage launches the mitochondrial amplification loop, leading to mitochondrial outer membrane permeabilization, cytochrome c release, apoptosome formation, and caspase-3 activation, resulting in apoptosis. Barbara Geering et al. Blood 2011;117:5953-5962 ©2011 by American Society of Hematology