Volume 118, Issue 4, Pages (April 2000)

Slides:



Advertisements
Similar presentations
Volume 131, Issue 1, Pages (July 2006)
Advertisements

Volume 119, Issue 5, Pages (November 2000)
IL-18 Downregulates Collagen Production in Human Dermal Fibroblasts via the ERK Pathway  Hee Jung Kim, Seok Bean Song, Jung Min Choi, Kyung Moon Kim,
Potent Mitogenicity of the RET/PTC3 Oncogene Correlates with Its Prevalence in Tall- Cell Variant of Papillary Thyroid Carcinoma  Fulvio Basolo, Riccardo.
Volume 114, Issue 4, Pages (April 1998)
Volume 109, Issue 2, Pages (April 2002)
A novel SHP-1/Grb2–dependent mechanism of negative regulation of cytokine-receptor signaling: contribution of SHP-1 C-terminal tyrosines in cytokine signaling.
W. L. Parker, M. D. , Ph. D. , K. W. Finnson, Ph. D. , H. Soe-Lin, B
Volume 9, Issue 5, Pages (November 1998)
Volume 131, Issue 1, Pages (July 2006)
Roles of the cytoplasmic domains of the α and β subunits of human granulocyte- macrophage colony-stimulating factor receptor  Akihiko Muto, PhDa, Sumiko.
Volume 118, Issue 4, Pages (April 2000)
PTF1α/p48 and cell proliferation
Volume 35, Issue 3, Pages (August 2009)
Requirement of heat shock protein 90 in mesangial cell mitogenesis
Volume 125, Issue 3, Pages (September 2003)
Volume 120, Issue 1, Pages (January 2001)
John F. Öhd, Katarina Wikström, Anita Sjölander  Gastroenterology 
Naoko Kanda, Shinichi Watanabe  Journal of Investigative Dermatology 
Volume 60, Issue 5, Pages (November 2001)
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,
IFN-γ Upregulates Expression of the Mouse Complement C1rA Gene in Keratinocytes via IFN-Regulatory Factor-1  Sung June Byun, Ik-Soo Jeon, Hyangkyu Lee,
Suppression of fibroblast growth factor receptor signaling inhibits pancreatic cancer growth in vitro and in vivo  Markus Wagner, Martha E. Lopez, Mitch.
Bone morphogenetic protein-2 induces apoptosis in human myeloma cells with modulation of STAT3 by Chiharu Kawamura, Masahiro Kizaki, Kenji Yamato, Hideo.
Arginine Methylation of STAT1 Modulates IFNα/β-Induced Transcription
Volume 138, Issue 5, Pages e2 (May 2010)
Volume 122, Issue 4, Pages (April 2002)
Volume 115, Issue 4, Pages (October 1998)
Volume 64, Issue 2, Pages (August 2003)
Decreased Growth Inhibitory Responses of Squamous Carcinoma Cells to Interferon-γ Involve Failure to Recruit cki Proteins into cdk2 Complexes  Beth L.
Akio Horiguchi, Mototsugu Oya, Ken Marumo, Masaru Murai 
Yongli Bai, Chun Yang, Kathrin Hu, Chris Elly, Yun-Cai Liu 
Volume 119, Issue 5, Pages (November 2000)
MUC1 Oncoprotein Stabilizes and Activates Estrogen Receptor α
Volume 119, Issue 2, Pages (August 2000)
Volume 59, Issue 5, Pages (May 2001)
Volume 58, Issue 3, Pages (September 2000)
1,25-dihydroxyvitamin D3 inhibits renal interstitial myofibroblast activation by inducing hepatocyte growth factor expression  Yingjian Li, Bradley C.
Volume 35, Issue 3, Pages (August 2009)
MUC1 Oncoprotein Stabilizes and Activates Estrogen Receptor α
Histamine Inhibits the Production of Interferon-induced Protein of 10 kDa in Human Squamous Cell Carcinoma and Melanoma  Naoko Kanda, Shinichi Watanabe 
Naoko Kanda, Shinichi Watanabe  Journal of Investigative Dermatology 
Volume 15, Issue 5, Pages (November 2001)
Cyclooxygenase-2 Inhibitor Enhances Whereas Prostaglandin E2Inhibits the Production of Interferon-Induced Protein of 10 kDa in Epidermoid Carcinoma A431 
Keratinocyte growth factor promotes goblet cell differentiation through regulation of goblet cell silencer inhibitor  Dai Iwakiri, Daniel K. Podolsky 
Volume 126, Issue 7, Pages (June 2004)
Volume 138, Issue 4, Pages (April 2010)
Interleukin-6-Resistant Melanoma Cells Exhibit Reduced Activation of STAT3 and Lack of Inhibition of Cyclin E-Associated Kinase Activity  Markus Böhm,
Volume 61, Issue 6, Pages (June 2002)
Volume 116, Issue 6, Pages (June 1999)
Activation of PPARγ leads to inhibition of anchorage-independent growth of human colorectal cancer cells  Jeffrey A. Brockman, Rajnish A. Gupta, Raymond.
Volume 39, Issue 3, Pages (September 2003)
Volume 127, Issue 4, Pages (October 2004)
Roles of the cytoplasmic domains of the α and β subunits of human granulocyte- macrophage colony-stimulating factor receptor  Akihiko Muto, PhDa, Sumiko.
Green Tea Polyphenol Epigallocatechin-3-Gallate Suppresses Collagen Production and Proliferation in Keloid Fibroblasts via Inhibition of the STAT3-Signaling.
IL-18 Downregulates Collagen Production in Human Dermal Fibroblasts via the ERK Pathway  Hee Jung Kim, Seok Bean Song, Jung Min Choi, Kyung Moon Kim,
Volume 119, Issue 1, Pages (July 2000)
Volume 123, Issue 6, Pages (December 2002)
The Prolyl Isomerase Pin1 Functions in Mitotic Chromosome Condensation
Interferon-γ-Mediated Growth Regulation of Melanoma Cells: Involvement of STAT1- Dependent and STAT1-Independent Signals  Anja Bosserhoff  Journal of Investigative.
Volume 122, Issue 1, Pages (January 2002)
Volume 67, Issue 6, Pages (June 2005)
Silva H Hanissian, Raif S Geha  Immunity 
Transcriptional Repression of miR-34 Family Contributes to p63-Mediated Cell Cycle Progression in Epidermal Cells  Dario Antonini, Monia T. Russo, Laura.
Volume 60, Issue 3, Pages (September 2001)
Volume 119, Issue 5, Pages (November 2000)
Volume 55, Issue 2, Pages (February 1999)
Naoko Kanda, Shinichi Watanabe  Journal of Investigative Dermatology 
Ultraviolet-B-Induced G1 Arrest is Mediated by Downregulation of Cyclin-Dependent Kinase 4 in Transformed Keratinocytes Lacking Functional p53  Arianna.
A. A. Honokiol inhibits TNF-induced NF-κB activation, IκBα phosphorylation, and IκBα degradation. Honokiol inhibits TNF-induced activation of NF-κB. H1299.
Presentation transcript:

Volume 118, Issue 4, Pages 735-748 (April 2000) Molecular mechanism of interferon alfa–Mediated growth inhibition in human neuroendocrine tumor cells  Katharina M. Detjen, Martina Welzel, Katrin Farwig, Felix H. Brembeck, Astrid Kaiser, Ernst-Otto Riecken, Bertram Wiedenmann, Stefan Rosewicz  Gastroenterology  Volume 118, Issue 4, Pages 735-748 (April 2000) DOI: 10.1016/S0016-5085(00)70143-0 Copyright © 2000 American Gastroenterological Association Terms and Conditions

Fig. 1 NE tumor cells express IFN-α receptor mRNA. Total RNA was extracted from QGP1, BON, and Capan1 cell lines, reverse-transcribed, and amplified by PCR using primers directed against the type I IFN receptor α chain. For size determination of the obtained PCR fragment, a 100-bp DNA standard was used (lane 1). Alternating sample lanes represent reactions with (+) or without (−) addition of reverse transcriptase. Gastroenterology 2000 118, 735-748DOI: (10.1016/S0016-5085(00)70143-0) Copyright © 2000 American Gastroenterological Association Terms and Conditions

Fig. 2 IFN-α treatment initiates activation of receptor-associated kinases in human NE tumor cells. Immunoblots of Jak1 and Tyk2 immunoprecipitates analyzing IFN-α–induced changes in the phosphotyrosine content of IFN-α receptor–associated kinases in (A) QGP1 and (B) BON cells. Cells were incubated with 1000 IU/mL IFN-α for the indicated periods. Blots were subsequently stripped and reprobed with antibodies against Jak1 or Tyk2 to confirm that equal amounts of immunocomplexes were evaluated. Because Jak1 phosphotyrosine signals were notably weaker in BON cells than in QGP1 cells, the autoradiography had to be exposed for 30 minutes compared with 1–2-minute exposures required in QGP1 cells. Gastroenterology 2000 118, 735-748DOI: (10.1016/S0016-5085(00)70143-0) Copyright © 2000 American Gastroenterological Association Terms and Conditions

Fig. 2 IFN-α treatment initiates activation of receptor-associated kinases in human NE tumor cells. Immunoblots of Jak1 and Tyk2 immunoprecipitates analyzing IFN-α–induced changes in the phosphotyrosine content of IFN-α receptor–associated kinases in (A) QGP1 and (B) BON cells. Cells were incubated with 1000 IU/mL IFN-α for the indicated periods. Blots were subsequently stripped and reprobed with antibodies against Jak1 or Tyk2 to confirm that equal amounts of immunocomplexes were evaluated. Because Jak1 phosphotyrosine signals were notably weaker in BON cells than in QGP1 cells, the autoradiography had to be exposed for 30 minutes compared with 1–2-minute exposures required in QGP1 cells. Gastroenterology 2000 118, 735-748DOI: (10.1016/S0016-5085(00)70143-0) Copyright © 2000 American Gastroenterological Association Terms and Conditions

Fig. 3 IFN-α time-dependently induces tyrosine phosphorylation of Stat1 and Stat2 in NE tumor cells. Both tyrosine phosphorylated (upper row) and total (lower row). Stat complement of Stat1 (upper panel) and Stat2 immunoprecipitates (lower panel) from IFN-α–treated cells were evaluated by immunoblotting. Cells were treated with 1000 IU/mL IFN-α for the indicated intervals. p91 and p84 indicate Stat 1 α- and β-splice variants, respectively, based on their molecular weight. Data are from representative experiments in (A) QGP1 and (B) BON cells. Gastroenterology 2000 118, 735-748DOI: (10.1016/S0016-5085(00)70143-0) Copyright © 2000 American Gastroenterological Association Terms and Conditions

Fig. 4 IFN-α stimulation results in transactivation of an ISRE-driven reporter construct. (A) Dose-dependent increase of relative luciferase activity (RLU) in IFN-α–treated BON cells transiently transfected with pGL2-ISRE-luc, but not mutant pGL2-ΔISRE-luc. Data represent means ± SEM from at least 3 separate experiments conducted in triplicate. (B) Immunoblot analysis of Stat1 and Stat2 in nuclear extracts of QGP1 cells, confirming the nuclear translocation of Stat transcription factors in cells stimulated with 1000 IU/mL IFN-α for the indicated times. Gastroenterology 2000 118, 735-748DOI: (10.1016/S0016-5085(00)70143-0) Copyright © 2000 American Gastroenterological Association Terms and Conditions

Fig. 5 IFN-α inhibits growth of NE tumor cells in a time- and dose-dependent manner. Subconfluent cells were treated with (A and B) 1000 IU/mL IFN-α (●) or vehicle (○) for the indicated time periods or with (C and D) 100 (□), 500 (■), and 1000 (▨) IU/mL for the indicated times. Viable cells were then manually counted in a hemacytometer. Data represent means ± SEM from at least 3–4 separate experiments, each conducted in triplicate. *P ≤ 0.05 compared with vehicle-treated controls. Gastroenterology 2000 118, 735-748DOI: (10.1016/S0016-5085(00)70143-0) Copyright © 2000 American Gastroenterological Association Terms and Conditions

Fig. 5 IFN-α inhibits growth of NE tumor cells in a time- and dose-dependent manner. Subconfluent cells were treated with (A and B) 1000 IU/mL IFN-α (●) or vehicle (○) for the indicated time periods or with (C and D) 100 (□), 500 (■), and 1000 (▨) IU/mL for the indicated times. Viable cells were then manually counted in a hemacytometer. Data represent means ± SEM from at least 3–4 separate experiments, each conducted in triplicate. *P ≤ 0.05 compared with vehicle-treated controls. Gastroenterology 2000 118, 735-748DOI: (10.1016/S0016-5085(00)70143-0) Copyright © 2000 American Gastroenterological Association Terms and Conditions

Fig. 5 IFN-α inhibits growth of NE tumor cells in a time- and dose-dependent manner. Subconfluent cells were treated with (A and B) 1000 IU/mL IFN-α (●) or vehicle (○) for the indicated time periods or with (C and D) 100 (□), 500 (■), and 1000 (▨) IU/mL for the indicated times. Viable cells were then manually counted in a hemacytometer. Data represent means ± SEM from at least 3–4 separate experiments, each conducted in triplicate. *P ≤ 0.05 compared with vehicle-treated controls. Gastroenterology 2000 118, 735-748DOI: (10.1016/S0016-5085(00)70143-0) Copyright © 2000 American Gastroenterological Association Terms and Conditions

Fig. 5 IFN-α inhibits growth of NE tumor cells in a time- and dose-dependent manner. Subconfluent cells were treated with (A and B) 1000 IU/mL IFN-α (●) or vehicle (○) for the indicated time periods or with (C and D) 100 (□), 500 (■), and 1000 (▨) IU/mL for the indicated times. Viable cells were then manually counted in a hemacytometer. Data represent means ± SEM from at least 3–4 separate experiments, each conducted in triplicate. *P ≤ 0.05 compared with vehicle-treated controls. Gastroenterology 2000 118, 735-748DOI: (10.1016/S0016-5085(00)70143-0) Copyright © 2000 American Gastroenterological Association Terms and Conditions

Fig. 6 IFN-α inhibits anchorage-independent growth of NE tumor cells. A single cell agar supension of (A) QGP1 or (B) BON cells was incubated with the indicated doses of IFN-α for 10 days, and colony formation was evaluated. Data represent means ± SEM from at least 3 separate experiments, each conducted in triplicate. *P ≤ 0.05 and **P ≤ 0.01 compared with vehicle-treated controls. Gastroenterology 2000 118, 735-748DOI: (10.1016/S0016-5085(00)70143-0) Copyright © 2000 American Gastroenterological Association Terms and Conditions

Fig. 7 IFN-α treatment does not affect Rb phosphorylation in NE tumor cells. (A) Immunoblot analysis of Rb expression and phosphorylation status in control and IFN-α–stimulated NE tumor cells as well as Capan1 exocrine pancreatic cancer cells. In each lane, 15 μg of whole cell lysates was separated by 7.5% SDS-PAGE. (B) Aliquots of the same lysates immunoblotted for expression of the CKI p16ink4a. Gastroenterology 2000 118, 735-748DOI: (10.1016/S0016-5085(00)70143-0) Copyright © 2000 American Gastroenterological Association Terms and Conditions

Fig. 8 IFN-α–treated NE tumor cells accumulate in the S phase of the cell cycle. (A) Representative fluorescence-activated cell sorter analysis showing changes in cell cycle distribution of QGP1 cells treated with 1000 IU/mL IFN-α for 48 hours. (B) Summary of IFN-α–induced cell cycle redistribution in QGP1 and BON cells. Data are expressed as mean ± SEM of the percentage of cells in the S phase (closed symbols) and the G1 phase (open symbols) compared with untreated control cultures, determined in 3 independent experiments. *P ≤ 0.05 and **P ≤ 0.01 compared with control. Gastroenterology 2000 118, 735-748DOI: (10.1016/S0016-5085(00)70143-0) Copyright © 2000 American Gastroenterological Association Terms and Conditions

Fig. 8 IFN-α–treated NE tumor cells accumulate in the S phase of the cell cycle. (A) Representative fluorescence-activated cell sorter analysis showing changes in cell cycle distribution of QGP1 cells treated with 1000 IU/mL IFN-α for 48 hours. (B) Summary of IFN-α–induced cell cycle redistribution in QGP1 and BON cells. Data are expressed as mean ± SEM of the percentage of cells in the S phase (closed symbols) and the G1 phase (open symbols) compared with untreated control cultures, determined in 3 independent experiments. *P ≤ 0.05 and **P ≤ 0.01 compared with control. Gastroenterology 2000 118, 735-748DOI: (10.1016/S0016-5085(00)70143-0) Copyright © 2000 American Gastroenterological Association Terms and Conditions

Fig. 9 IFN-α treatment does not impair CDK2 activity in randomly cycling QGP1 cells. (A) Effects of IFN-α on expression of cyclin E and on CDK2 activity as determined by immunoblotting and histone H1–kinase assays, respectively. (B) Immunoblot analysis to determine IFN-α–dependent regulation of S–G2/M phase cyclin expression (cyclin A and B, upper panel) and determination of histone H1–kinase activity of CDC2 complexes prepared from randomly cycling cells (lower panel). Immunoblots and kinase assays are representative of at least 2–3 experiments. Gastroenterology 2000 118, 735-748DOI: (10.1016/S0016-5085(00)70143-0) Copyright © 2000 American Gastroenterological Association Terms and Conditions

Fig. 10 IFN-α treatment delays S-phase progression in synchronized NE tumor cells. Serum-deprived QGP1 cells synchronized in G0/G1 were stimulated by addition of 10% FCS to reenter the cell cycle either in the absence (upper panel) or presence (lower panel) of 1000 IU/mL IFN-α. (A) Cells were harvested at the indicated time points after readdition of serum, and cell cycle distribution was evaluated by flow cytometry. (B) Cell cycle distribution of 4–7 independent experiments was quantitated, and the fraction of IFN-α–treated cells in S and G2/M phase is given as percentage of time-matched, untreated control cultures. *P ≤ 0.05 compared with time-matched controls. Gastroenterology 2000 118, 735-748DOI: (10.1016/S0016-5085(00)70143-0) Copyright © 2000 American Gastroenterological Association Terms and Conditions

Fig. 10 IFN-α treatment delays S-phase progression in synchronized NE tumor cells. Serum-deprived QGP1 cells synchronized in G0/G1 were stimulated by addition of 10% FCS to reenter the cell cycle either in the absence (upper panel) or presence (lower panel) of 1000 IU/mL IFN-α. (A) Cells were harvested at the indicated time points after readdition of serum, and cell cycle distribution was evaluated by flow cytometry. (B) Cell cycle distribution of 4–7 independent experiments was quantitated, and the fraction of IFN-α–treated cells in S and G2/M phase is given as percentage of time-matched, untreated control cultures. *P ≤ 0.05 compared with time-matched controls. Gastroenterology 2000 118, 735-748DOI: (10.1016/S0016-5085(00)70143-0) Copyright © 2000 American Gastroenterological Association Terms and Conditions

Fig. 11 IFN-α–treated QGP1 cells fail to induce cyclin B, resulting in reduced CDC2 activity. (A) Whole-cell lysates of synchronized QGP1 cells were investigated for cyclin B expression by immunoblotting under control conditions or in the presence of 1000 IU/mL IFN-α. (B) Lysates from the same experiment were also used to immunoprecipitate CDC2 and subsequently assess CDC2-associated histone kinase activity. (C) Cyclin B expression (□) and CDC2 activity (■) were quantitated and the signal intensity was expressed as a percentage of the time matched, untreated controls. Results are representative of 3 independent experiments with similar results. Gastroenterology 2000 118, 735-748DOI: (10.1016/S0016-5085(00)70143-0) Copyright © 2000 American Gastroenterological Association Terms and Conditions

Fig. 11 IFN-α–treated QGP1 cells fail to induce cyclin B, resulting in reduced CDC2 activity. (A) Whole-cell lysates of synchronized QGP1 cells were investigated for cyclin B expression by immunoblotting under control conditions or in the presence of 1000 IU/mL IFN-α. (B) Lysates from the same experiment were also used to immunoprecipitate CDC2 and subsequently assess CDC2-associated histone kinase activity. (C) Cyclin B expression (□) and CDC2 activity (■) were quantitated and the signal intensity was expressed as a percentage of the time matched, untreated controls. Results are representative of 3 independent experiments with similar results. Gastroenterology 2000 118, 735-748DOI: (10.1016/S0016-5085(00)70143-0) Copyright © 2000 American Gastroenterological Association Terms and Conditions

Fig. 11 IFN-α–treated QGP1 cells fail to induce cyclin B, resulting in reduced CDC2 activity. (A) Whole-cell lysates of synchronized QGP1 cells were investigated for cyclin B expression by immunoblotting under control conditions or in the presence of 1000 IU/mL IFN-α. (B) Lysates from the same experiment were also used to immunoprecipitate CDC2 and subsequently assess CDC2-associated histone kinase activity. (C) Cyclin B expression (□) and CDC2 activity (■) were quantitated and the signal intensity was expressed as a percentage of the time matched, untreated controls. Results are representative of 3 independent experiments with similar results. Gastroenterology 2000 118, 735-748DOI: (10.1016/S0016-5085(00)70143-0) Copyright © 2000 American Gastroenterological Association Terms and Conditions