1
|
Myacheva K, Walsh A, Riester M, Pelos G, Carl J, Diederichs S. CRISPRi screening identifies CASP8AP2 as an essential viability factor in lung cancer controlling tumor cell death via the AP-1 pathway. Cancer Lett 2023; 552:215958. [PMID: 36252816 DOI: 10.1016/j.canlet.2022.215958] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/07/2022] [Accepted: 10/07/2022] [Indexed: 02/09/2023]
Abstract
Since lung cancer remains the leading cause of cancer death globally, there is an urgent demand for novel therapeutic targets. We carried out a CRISPR interference (CRISPRi) loss-of-function screen for human lung adenocarcinoma (LUAD) targeting 2098 deregulated genes using a customized algorithm to comprehensively probe the functionality of every resolvable transcriptional start site (TSS). CASP8AP2 was identified as the only hit that significantly affected the viability of all eight screened LUAD cell lines while the viability of non-transformed lung cells was only moderately impacted. Knockdown (KD) of CASP8AP2 induced both autophagy and apoptotic cell death pathways. Systematic expression profiling linked the AP-1 transcription factor to the CASP8AP2 KD-induced cancer cell death. Furthermore, inhibition of AP-1 reverted the CASP8AP2 silencing-induced phenotype. Overall, the tailored CRISPRi screen profiled the impact of over 2000 genes on the survival of eight LUAD cell lines and identified the CASP8AP2 - AP-1 axis mediating lung cancer viability.
Collapse
Affiliation(s)
- Ksenia Myacheva
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) - Partner Site Freiburg, Germany; Division of RNA Biology & Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andrew Walsh
- siTOOLs Biotech GmbH, Lochhamerstr. 29A, Planegg, Martinsried, Germany
| | - Marisa Riester
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) - Partner Site Freiburg, Germany
| | - Giulia Pelos
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) - Partner Site Freiburg, Germany
| | - Jane Carl
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) - Partner Site Freiburg, Germany
| | - Sven Diederichs
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) - Partner Site Freiburg, Germany; Division of RNA Biology & Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
2
|
Omega-3 fatty acid protects cardiomyocytes against hypoxia-induced injury through targeting MiR-210-3p/CASP8AP2 axis. Mol Cell Biochem 2021; 476:2999-3007. [PMID: 33791918 DOI: 10.1007/s11010-021-04141-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 03/19/2021] [Indexed: 10/21/2022]
Abstract
MicroRNAs (miRs) regulate diverse biological functions in both normal and pathological cellular conditions by post-transcriptional regulation of various genes expression. Nevertheless, the role of miRs in regulating the protective functions of omega-3 fatty acid in relation to hypoxia in cardiomyocytes remains unknown. The aim of this study was to investigate the effects of omega-3 fatty acid supplementation on cardiomyocyte apoptosis and further delineate the mechanisms underlying microRNA-210 (miRNA-210)-induced cardiomyocyte apoptosis in vitro. H9C2 cultured cells were first subjected to hypoxia followed by a subsequent treatment with main component of the Omega-3 fatty acid, Docosahexaenoic Acid (DHA). Cell apoptosis were detected by flow cytometry and the expression of miR-210-3p were detected by RT-qPCR and caspase-8-associated protein 2 (CASP8AP2) at protein levels by immunoblotting. Dual luciferase assay was used to verify the mutual effect between miR-210-3p and the 3'-untranslated region (UTR) of CASP8AP2 gene. DHA was shown to reduce apoptosis in H9C2 cells subjected to hypoxia. While DHA caused a significant increase in the expression of miR-210-3p, there was a marked reduction in the protein expression of CASP8AP2. MiR-210-3p and CASP8AP2 were significantly increased in H9C2 cardiomyocyte subjected to hypoxia. Overexpression of miR-210-3p could ameliorate hypoxia-induced apoptosis in H9C2 cells. MiR-210-3p negatively regulated CASP8AP2 expression at the transcriptional level. Both miR-210-3p mimic and CASP8AP2 siRNA could efficiently inhibit apoptosis in H9C2 cardiomyocyte subjected to hypoxia. We provide strong evidence showing that Omega-3 fatty acids can attenuate apoptosis in cardiomyocyte under hypoxic conditions via the up-regulation of miR-210-3p and targeting CASP8AP2 signaling pathway.
Collapse
|
3
|
Minamida Y, Someda M, Yonehara S. FLASH/casp8ap2 is indispensable for early embryogenesis but dispensable for proliferation and differentiation of ES cells. PLoS One 2014; 9:e108032. [PMID: 25238250 PMCID: PMC4169604 DOI: 10.1371/journal.pone.0108032] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 08/24/2014] [Indexed: 11/19/2022] Open
Abstract
FLICE/caspase-8-associated huge protein (FLASH)/casp8ap2 is involved in various cellular functions, such as cell cycle progression, transcriptional regulation, the regulation of apoptosis, and the regulation of histone gene expression. The down-regulated expression of FLASH has been shown to inhibit cell cycle progression in the S phase in many kinds of mice and human cell lines and the inhibition of cell cycle progression may be attributed to the suppressed expression of replication-dependent histone genes. We here demonstrated that the induced knockout of FLASH never affected cell cycle progression in ES cells, in which the expression of core histone genes was decreased to levels similar to those in human KB cells sensitive to the knockdown of FLASH. In addition, the FLASH conditional knockout ES cells could differentiate normally into not only mesodermal and endodermal cells, but also trophoblasts. In order to investigate the function of FLASH in early embryogenesis in vivo, we also examined a FLASH mutant mouse, in which FLASH mutant allele did not express FLASH mRNA in embryos and most adult organs, except for the testis. FLASH mutant embryos died between E3.5 and E8.5. Furthermore, the in vitro cultivation of FLASH mutant embryos generated by in vitro fertilization showed embryonic lethality at the pre-implantation stage by inhibiting the hatching of embryos and their adherence to substrates. Taken together, these results indicate that FLASH plays an important role in early embryogenesis, but is not essential for either the proliferation or differentiation of ES cells.
Collapse
Affiliation(s)
- Yoshitaka Minamida
- Laboratory of Molecular and Cellular Biology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe, Sakyo, Kyoto, Japan
| | - Masataka Someda
- Laboratory of Molecular and Cellular Biology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe, Sakyo, Kyoto, Japan
| | - Shin Yonehara
- Laboratory of Molecular and Cellular Biology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe, Sakyo, Kyoto, Japan
- * E-mail:
| |
Collapse
|
4
|
Juárez-Velázquez R, Reyes-León A, Salas-Labadía C, Rivera-Luna R, Velasco-Hidalgo L, López-Hernández G, López-Santiago N, Paredes-Aguilera R, Domínguez-López A, Bernáldez R, Pérez-Vera P. Significance of CASP8AP2 and H2AFZ expression in survival and risk of relapse in children with acute lymphoblastic leukemia. Leuk Lymphoma 2014; 55:2305-11. [PMID: 24397596 DOI: 10.3109/10428194.2013.878458] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Novel biomarkers for risk refinement and stratification in childhood acute lymphoblastic leukemia (ALL) are needed to optimize treatment results. We studied the expression of CASP8AP2 and H2AFZ associated with relapse and survival in bone marrow samples from newly diagnosed children with ALL. We found: (a) an increased risk for early relapse in those patients with low expression of CASP8AP2 (odds ratio [OR] 3.93, 95% confidence interval [CI] 1.40-11.02, p < 0.05) confirming its usefulness as a predictive risk marker, although H2AFZ did not present the same effect; (b) patients with low expressions of CASP8AP2 and H2AFZ had inferior survival rates (p < 0.001); (c) the predictive values regarding low expressions of H2AFZ and CASP8AP2 and high white blood cell count suggest that these features could help to identify more accurately patients at greater risk of relapse.
Collapse
Affiliation(s)
- Rocío Juárez-Velázquez
- Laboratorio de Cultivo de Tejidos, Instituto Nacional de Pediatría , México D.F. , México
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Vennemann A, Hofmann TG. SUMO regulates proteasome-dependent degradation of FLASH/Casp8AP2. Cell Cycle 2013; 12:1914-21. [PMID: 23673342 DOI: 10.4161/cc.24943] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
FLASH/Casp8AP2 is a huge multifunctional protein involved in multiple cellular processes, reaching from death receptor signaling to regulation of histone gene transcription and histone mRNA processing. Previous work has shown that FLASH localizes to Cajal bodies and promyelocytic leukemia (PML) bodies. However, the function of its nuclear body association remains unclear. Here we demonstrate that murine FLASH is covalently modified by SUMO at Lys residue 1792. Interestingly, ectopic expression of SUMO results in proteasome-dependent degradation of FLASH. A point mutant of FLASH with a mutated SUMO acceptor lysine residue, FLASH(K1792R), is resistant to SUMO-induced degradation. Finally, we show that arsenic trioxide, a drug known to potentiate SUMO modification and degradation of PML, triggers recruitment of FLASH to PML bodies and concomitant loss of FLASH protein. Our data suggest that SUMO targets FLASH for proteasome-dependent degradation, which is associated with recruitment of FLASH to PML bodies.
Collapse
Affiliation(s)
- Astrid Vennemann
- German Cancer Research Center (DKFZ), Research Group Cellular Senescence, DKFZ-ZMBH Alliance, Heidelberg, Germany
| | | |
Collapse
|
6
|
Chen S, Evans HG, Evans DR. FLASH knockdown sensitizes cells to Fas-mediated apoptosis via down-regulation of the anti-apoptotic proteins, MCL-1 and Cflip short. PLoS One 2012; 7:e32971. [PMID: 22427918 PMCID: PMC3302898 DOI: 10.1371/journal.pone.0032971] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 02/06/2012] [Indexed: 12/24/2022] Open
Abstract
FLASH (FLICE-associated huge protein or CASP8AP2) is a large multifunctional protein that is involved in many cellular processes associated with cell death and survival. It has been reported to promote apoptosis, but we show here that depletion of FLASH in HT1080 cells by siRNA interference can also accelerate the process. As shown previously, depletion of FLASH halts growth by down-regulating histone biosynthesis and arrests the cell cycle in S-phase. FLASH knockdown followed by stimulating the cells with Fas ligand or anti-Fas antibodies was found to be associated with a more rapid cleavage of PARP, accelerated activation of caspase-8 and the executioner caspase-3 and rapid progression to cellular disintegration. As is the case for most anti-apoptotic proteins, FLASH was degraded soon after the onset of apoptosis. Depletion of FLASH also resulted in the reduced intracellular levels of the anti-apoptotic proteins, MCL-1 and the short isoform of cFLIP. FLASH knockdown in HT1080 mutant cells defective in p53 did not significantly accelerate Fas mediated apoptosis indicating that the effect was dependent on functional p53. Collectively, these results suggest that under some circumstances, FLASH suppresses apoptosis.
Collapse
Affiliation(s)
- Song Chen
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Hedeel Guy Evans
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Department of Chemistry, Eastern Michigan University, Ypsilanti, Michigan, United States of America
| | - David R. Evans
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- * E-mail:
| |
Collapse
|
7
|
Hummon AB, Pitt JJ, Camps J, Emons G, Skube SB, Huppi K, Jones TL, Beissbarth T, Kramer F, Grade M, Difilippantonio MJ, Ried T, Caplen NJ. Systems-wide RNAi analysis of CASP8AP2/FLASH shows transcriptional deregulation of the replication-dependent histone genes and extensive effects on the transcriptome of colorectal cancer cells. Mol Cancer 2012; 11:1. [PMID: 22216762 PMCID: PMC3281783 DOI: 10.1186/1476-4598-11-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 01/04/2012] [Indexed: 11/11/2022] Open
Abstract
Background Colorectal carcinomas (CRC) carry massive genetic and transcriptional alterations that influence multiple cellular pathways. The study of proteins whose loss-of-function (LOF) alters the growth of CRC cells can be used to further understand the cellular processes cancer cells depend upon for survival. Results A small-scale RNAi screen of ~400 genes conducted in SW480 CRC cells identified several candidate genes as required for the viability of CRC cells, most prominently CASP8AP2/FLASH. To understand the function of this gene in maintaining the viability of CRC cells in an unbiased manner, we generated gene specific expression profiles following RNAi. Silencing of CASP8AP2/FLASH resulted in altered expression of over 2500 genes enriched for genes associated with cellular growth and proliferation. Loss of CASP8AP2/FLASH function was significantly associated with altered transcription of the genes encoding the replication-dependent histone proteins as a result of the expression of the non-canonical polyA variants of these transcripts. Silencing of CASP8AP2/FLASH also mediated enrichment of changes in the expression of targets of the NFκB and MYC transcription factors. These findings were confirmed by whole transcriptome analysis of CASP8AP2/FLASH silenced cells at multiple time points. Finally, we identified and validated that CASP8AP2/FLASH LOF increases the expression of neurofilament heavy polypeptide (NEFH), a protein recently linked to regulation of the AKT1/ß-catenin pathway. Conclusions We have used unbiased RNAi based approaches to identify and characterize the function of CASP8AP2/FLASH, a protein not previously reported as required for cell survival. This study further defines the role CASP8AP2/FLASH plays in the regulating expression of the replication-dependent histones and shows that its LOF results in broad and reproducible effects on the transcriptome of colorectal cancer cells including the induction of expression of the recently described tumor suppressor gene NEFH.
Collapse
Affiliation(s)
- Amanda B Hummon
- Gene Silencing Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Burch BD, Godfrey AC, Gasdaska PY, Salzler HR, Duronio RJ, Marzluff WF, Dominski Z. Interaction between FLASH and Lsm11 is essential for histone pre-mRNA processing in vivo in Drosophila. RNA (NEW YORK, N.Y.) 2011; 17:1132-47. [PMID: 21525146 PMCID: PMC3096045 DOI: 10.1261/rna.2566811] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Metazoan replication-dependent histone mRNAs are the only nonpolyadenylated cellular mRNAs. Formation of the histone mRNA 3' end requires the U7 snRNP, which contains Lsm10 and Lsm11, and FLASH, a processing factor that binds Lsm11. Here, we identify sequences in Drosophila FLASH (dFLASH) that bind Drosophila Lsm11 (dLsm11), allow localization of dFLASH to the nucleus and histone locus body (HLB), and participate in histone pre-mRNA processing in vivo. Amino acids 105-154 of dFLASH bind to amino acids 1-78 of dLsm11. A two-amino acid mutation of dLsm11 that prevents dFLASH binding but does not affect localization of U7 snRNP to the HLB cannot rescue the lethality or histone pre-mRNA processing defects resulting from an Lsm11 null mutation. The last 45 amino acids of FLASH are required for efficient localization to the HLB in Drosophila cultured cells. Removing the first 64 amino acids of FLASH has no effect on processing in vivo. Removal of 13 additional amino acids of dFLASH results in a dominant negative protein that binds Lsm11 but inhibits processing of histone pre-mRNA in vivo. Inhibition requires the Lsm11 binding site, suggesting that the mutant dFLASH protein sequesters the U7 snRNP in an inactive complex and that residues between 64 and 77 of dFLASH interact with a factor required for processing. Together, these studies demonstrate that direct interaction between dFLASH and dLsm11 is essential for histone pre-mRNA processing in vivo and for proper development and viability in flies.
Collapse
MESH Headings
- Animals
- Binding Sites
- Carrier Proteins/chemistry
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cells, Cultured
- Drosophila/genetics
- Drosophila/metabolism
- Drosophila Proteins/chemistry
- Drosophila Proteins/genetics
- Drosophila Proteins/metabolism
- Histones/genetics
- Histones/metabolism
- RNA Precursors/metabolism
- RNA Processing, Post-Transcriptional
- RNA, Heterogeneous Nuclear/genetics
- RNA, Heterogeneous Nuclear/metabolism
- RNA, Messenger/metabolism
- Ribonucleoprotein, U7 Small Nuclear/genetics
- Ribonucleoprotein, U7 Small Nuclear/metabolism
- Ribonucleoproteins, Small Nuclear/chemistry
- Ribonucleoproteins, Small Nuclear/genetics
- Ribonucleoproteins, Small Nuclear/metabolism
Collapse
Affiliation(s)
- Brandon D Burch
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
Alm-Kristiansen AH, Lorenzo PI, Molværsmyr AK, Matre V, Ledsaak M, Sæther T, Gabrielsen OS. PIAS1 interacts with FLASH and enhances its co-activation of c-Myb. Mol Cancer 2011; 10:21. [PMID: 21338522 PMCID: PMC3050860 DOI: 10.1186/1476-4598-10-21] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2010] [Accepted: 02/21/2011] [Indexed: 11/15/2022] Open
Abstract
Background FLASH is a huge nuclear protein involved in various cellular functions such as apoptosis signalling, NF-κB activation, S-phase regulation, processing of histone pre-mRNAs, and co-regulation of transcription. Recently, we identified FLASH as a co-activator of the transcription factor c-Myb and found FLASH to be tightly associated with active transcription foci. As a huge multifunctional protein, FLASH is expected to have many interaction partners, some which may shed light on its function as a transcriptional regulator. Results To find additional FLASH-associated proteins, we performed a yeast two-hybrid (Y2H) screening with FLASH as bait and identified the SUMO E3 ligase PIAS1 as an interaction partner. The association appears to involve two distinct interaction surfaces in FLASH. We verified the interaction by Y2H-mating, GST pulldowns, co-IP and ChIP. FLASH and PIAS1 were found to co-localize in nuclear speckles. Functional assays revealed that PIAS1 enhances the intrinsic transcriptional activity of FLASH in a RING finger-dependent manner. Furthermore, PIAS1 also augments the specific activity of c-Myb, and cooperates with FLASH to further co-activate c-Myb. The three proteins, FLASH, PIAS1, and c-Myb, are all co-localized with active RNA polymerase II foci, resembling transcription factories. Conclusions We conclude that PIAS1 is a common partner for two cancer-related nuclear factors, c-Myb and FLASH. Our results point to a functional cooperation between FLASH and PIAS1 in the enhancement of c-Myb activity in active nuclear foci.
Collapse
|
10
|
Tanaka M, Kamitani T. Cytoplasmic relocation of Daxx induced by Ro52 and FLASH. Histochem Cell Biol 2010; 134:297-306. [PMID: 20697732 DOI: 10.1007/s00418-010-0734-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2010] [Indexed: 12/23/2022]
Abstract
The RING-finger protein Ro52/TRIM21 is known to be an autoantigen and is recognized by anti-Ro/SSA antibodies, which are commonly found in patients with Sjögren's syndrome and systemic lupus erythematosus. We recently showed that Ro52 is an E3 ubiquitin ligase and localizes to cytoplasmic bodies that are highly motile along the microtubule network. To expand our knowledge of Ro52, we searched partners co-operating with Ro52. We performed a yeast two-hybrid screening of a human brain cDNA library with Ro52 as bait. This screening identified several genes encoding Ro52-interacting proteins, including the apoptosis-related proteins, Daxx and FLASH. Further yeast two-hybrid assays revealed that Daxx binds to the B30.2 domain of Ro52 and that FLASH binds to coiled-coil domains of Ro52 through its death-effector domain-recruiting domain. These results suggest that Ro52, Daxx, and FLASH form heteromeric protein complexes. Indeed, this was supported by results of immunoprecipitation experiments in which we found that Daxx is co-immunoprecipitated with Ro52 in the presence of overexpressed FLASH. Importantly, our fluorescence microscopy revealed that, although Daxx is predominantly located in the nucleus, overexpression of both Ro52 and FLASH leads to relocation of Daxx into the cytoplasm. Thus, Ro52 seems to co-operate with FLASH to induce cytoplasmic localization of Daxx in cells.
Collapse
Affiliation(s)
- Makoto Tanaka
- Department of Medicine, Center for Molecular Chaperone/Radiobiology and Cancer Virology, MCG Cancer Center, Medical College of Georgia, 1410 Laney Walker Blvd., CN-3141, Augusta, GA 30912, USA
| | | |
Collapse
|
11
|
Ha H, Han D, Choi Y. TRAF-mediated TNFR-family signaling. CURRENT PROTOCOLS IN IMMUNOLOGY 2009; Chapter 11:11.9D.1-11.9D.19. [PMID: 19918944 DOI: 10.1002/0471142735.im1109ds87] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The tumor necrosis factor (TNF) superfamily consists of a wide variety of cell-bound and secreted proteins that regulate numerous cellular processes. In particular, TNF-family proteins regulate the proliferation and death of tumor cells, as well as activated immune cells. This overview discusses the mammalian TNF receptor-associated factors (TRAFs), of which TRAF1, 2, 3, 5, and 6 have been shown to interact directly or indirectly with members of the TNF receptor superfamily. Structural features of TRAF proteins are described along with a discussion of TRAF-interacting proteins and the signaling pathways activated by the TRAF proteins. Finally, we examine the phenotypes observed in TRAF-knockout mice.
Collapse
Affiliation(s)
- Hyunil Ha
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Daehee Han
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Yongwon Choi
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
12
|
Kang HJ, Lee SH, Price JE, Kim LS. Curcumin suppresses the paclitaxel-induced nuclear factor-kappaB in breast cancer cells and potentiates the growth inhibitory effect of paclitaxel in a breast cancer nude mice model. Breast J 2009; 15:223-9. [PMID: 19645775 DOI: 10.1111/j.1524-4741.2009.00709.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Most anticancer agents activate nuclear factor kappa B (NF-kappaB), which can mediate cell survival, proliferation, and metastasis. Curcumin has been shown to inhibit the growth of various cancer cells, without toxicity to normal cells. The antitumor effects of curcumin could be due in part to the inactivation of NF-kappaB. We hypothesize that blocking NF-kappaB activity may augment paclitaxel cancer chemotherapy. In this study, we investigated whether the inactivation of NF-kappaB by curcumin would enhance the efficacy of paclitaxel for inhibiting breast cancer growth in vitro and in vivo. We confirmed that curcumin inhibited paclitaxel-induced activation of NF-kappaB and potentiated the growth inhibitory effect of paclitaxel in MDA-MB-231 breast cancer cells. The combination of curcumin with paclitaxel elicited significantly greater inhibition of cell growth and more apoptosis, compared with either agent alone. In an experimental breast cancer murine model using MDA-MB-231 cells, combination therapy with paclitaxel and curcumin significantly reduced tumor size and decreased tumor cell proliferation, increased apoptosis, and decreased the expression of matrix metalloprotease 9 compared with either agent alone. These results clearly suggest that a curcumin-paclitaxel combination could be a novel strategy for the treatment of breast cancer.
Collapse
Affiliation(s)
- Hee Joon Kang
- Division of Breast & Endocrine Surgery, Hallym Sacred Heart Hospital, College of Medicine, Hallym University, Dongan-Gu, Anyang, Korea
| | | | | | | |
Collapse
|
13
|
Lukas TJ, Wang AL, Yuan M, Neufeld AH. Early cellular signaling responses to axonal injury. Cell Commun Signal 2009; 7:5. [PMID: 19284657 PMCID: PMC2661080 DOI: 10.1186/1478-811x-7-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2008] [Accepted: 03/13/2009] [Indexed: 12/20/2022] Open
Abstract
Background We have used optic nerve injury as a model to study early signaling events in neuronal tissue following axonal injury. Optic nerve injury results in the selective death of retinal ganglion cells (RGCs). The time course of cell death takes place over a period of days with the earliest detection of RGC death at about 48 hr post injury. We hypothesized that in the period immediately following axonal injury, there are changes in the soma that signal surrounding glia and neurons and that start programmed cell death. In the current study, we investigated early changes in cellular signaling and gene expression that occur within the first 6 hrs post optic nerve injury. Results We found evidence of cell to cell signaling within 30 min of axonal injury. We detected differences in phosphoproteins and gene expression within the 6 hrs time period. Activation of TNFα and glutamate receptors, two pathways that can initiate cell death, begins in RGCs within 6 hrs following axonal injury. Differential gene expression at 6 hrs post injury included genes involved in cytokine, neurotrophic factor signaling (Socs3) and apoptosis (Bax). Conclusion We interpret our studies to indicate that both neurons and glia in the retina have been signaled within 30 min after optic nerve injury. The signals are probably initiated by the RGC soma. In addition, signals activating cellular death pathways occur within 6 hrs of injury, which likely lead to RGC degeneration.
Collapse
Affiliation(s)
- Thomas J Lukas
- Forsythe Laboratory for the Investigation of the Aging Retina, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| | | | | | | |
Collapse
|
14
|
Poxvirus MC160 protein utilizes multiple mechanisms to inhibit NF-kappaB activation mediated via components of the tumor necrosis factor receptor 1 signal transduction pathway. J Virol 2009; 83:3162-74. [PMID: 19158250 DOI: 10.1128/jvi.02009-08] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Poxviruses express proteins that limit host immune responses to infection. For example, the molluscum contagiosum virus MC160 protein inhibits tumor necrosis factor alpha (TNF-alpha)-induced NF-kappaB activation. This event correlates with MC160-induced IKK1 protein degradation, suggesting a mechanism for the above-mentioned phenotype. IKK1 is stabilized when it associates with the cellular heat shock protein 90 (Hsp90). Here, Hsp90 overexpression restored IKK1 levels in MC160-expressing cells, suggesting that MC160 competitively interacted with Hsp90. In support of this, further investigation showed that a mutant MC160 protein comprising only the C-terminal region (C protein) immunoprecipitated with Hsp90. In contrast, Hsp90 IP with a mutant MC160 protein consisting of only the N-terminal tandem death effector domains (DEDs) (N protein) was dramatically decreased. Since cells expressing either the N or C mutant MC160 protein remained similarly resistant to TNF-alpha-induced NF-kappaB activation, the N mutant protein probably utilized a different mechanism for inhibiting NF-kappaB. One likely mechanism for the N protein lies in its association with the DED-containing procaspase-8 protein, a cellular apoptosis precursor protein that regulates NF-kappaB activation. Here, IPs revealed that this association relied on the presence of the DED-containing N terminus of the MC160 protein but not the C-terminal portion. These interactions appear to have relevance with NF-kappaB activation, since the expression of the viral DEDs strongly inhibited procaspase-8-mediated NF-kappaB activation, an event not substantially altered by the C protein. Thus, the MC160 protein utilizes at least two distinct mechanisms for impeding NF-kappaB activation, association with Hsp90 to result in IKK1 protein degradation or interaction with procaspase-8.
Collapse
|
15
|
|
16
|
Jeong EG, Lee SH, Yoo NJ, Lee SH. Mutational analysis of FLASH and PTPN13 genes in colorectal carcinomas. Pathology 2008; 40:31-4. [DOI: 10.1080/00313020701716441] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
17
|
Chung C, Park SG, Park YM, Joh JW, Jung G. Interferon-gamma sensitizes hepatitis B virus-expressing hepatocarcinoma cells to 5-fluorouracil through inhibition of hepatitis B virus-mediated nuclear factor-kappaB activation. Cancer Sci 2007; 98:1758-66. [PMID: 17711513 PMCID: PMC11159465 DOI: 10.1111/j.1349-7006.2007.00591.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2007] [Revised: 06/29/2007] [Accepted: 07/02/2007] [Indexed: 12/17/2022] Open
Abstract
Nuclear factor (NF)-kappaB is important for immune responses and cell survival; however, abnormal activation of NF-kappaB is linked with many types of diseases, including hepatocellular carcinoma (HCC). Our previous report indicated that hepatitis B virus (HBV) induces NF-kappaB activation through NF-kappaB-inducing kinase (NIK), and this can be blocked specifically by interferon (IFN)-gamma. In the present study, we report that HBV expression in HCC cell lines induces drug resistance against 5-fluorouracil (5-FU). This drug resistance was abolished by inhibition of NF-kappaB activation through small interfering RNA-mediated NIK 'knockdown' and IFN-gamma treatment. In addition to the reduced NF-kappaB activation and drug resistance, the upregulated growth arrest- and DNA damage-inducible protein 45beta (Gadd45beta) in HBV-expressing HCC cell lines was downregulated by the small interfering RNA-mediated NIK knockdown and IFN-gamma treatment. The overexpression of Gadd45beta in HCC cell lines also induces drug resistance against 5-FU. Based on our data, we suggest that IFN-gamma treatment might be helpful for chemotherapy in HBV-integrated HCC through inhibition of the NIK-mediated NF-kappaB activation and downregulation of the NF-kappaB target gene Gadd45beta.
Collapse
Affiliation(s)
- Chan Chung
- School of Biological Sciences, Seoul National University, Shillim-dong, Kwanak-gu, Seoul 151-742, Korea
| | | | | | | | | |
Collapse
|
18
|
Jeong EG, Lee SH, Lee HW, Soung YH, Yoo NJ, Lee SH. Immunohistochemical and mutational analysis of FLASH in gastric carcinomas. APMIS 2007; 115:900-5. [PMID: 17696945 DOI: 10.1111/j.1600-0463.2007.apm_706.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
FLASH was initially identified as a pro-apoptotic protein that transmits an apoptosis signal during death receptor-induced apoptosis. Additionally, diverse biologic roles of FLASH, including TNF-induced NF-kappaB activation, cell-cycle progression and cell division, have been identified. Although such functions are important in cancer pathogenesis, little is known about the alterations of FLASH gene and FLASH protein expression in human cancers. In this study, we analyzed the expression of FLASH protein in 60 gastric adenocarcinomas by immunohistochemistry. We furthermore analyzed mutation of FLASH in exon 8, where two polyadenine tracts ((A)8 and (A)9) are present, by single-strand conformation polymorphism (SSCP) assay in 184 gastric adenocarcinomas. By immunohistochemistry, FLASH protein expression in cancer cells was detected positively in 42 gastric carcinoma tissues (70%), whereas its expression in epithelial cells of normal gastric mucosa was shown as no or very weak intensity. Mutational analysis detected one FLASH mutation in the gastric carcinomas (0.5%). The increased expression of FLASH in the malignant gastric epithelial cells compared to the normal mucosal epithelial cells suggests that FLASH expression may play a role in gastric tumorigenesis. Also, the data suggest that somatic mutation of FLASH is a rare event in gastric carcinomas.
Collapse
Affiliation(s)
- Eun Goo Jeong
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
19
|
Ng MKC, Wu J, Chang E, Wang BY, Katzenberg-Clark R, Ishii-Watabe A, Cooke JP. A Central Role for Nicotinic Cholinergic Regulation of Growth Factor–Induced Endothelial Cell Migration. Arterioscler Thromb Vasc Biol 2007; 27:106-12. [PMID: 17082486 DOI: 10.1161/01.atv.0000251517.98396.4a] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE An endothelial nicotinic acetylcholine receptor (nAChR) participates in atherogenesis and tumorigenesis by promoting neovascularization. To date, the mechanisms of nAChR-mediated angiogenesis and their relationship to angiogenic factors, eg, VEGF and bFGF, are unknown. METHODS AND RESULTS Nicotine induced dose-dependent human microvascular endothelial cell (HMVEC) migration, a key angiogenesis event, to an extent which was equivalent in magnitude to bFGF (10 ng/mL) but less than for VEGF (10 ng/mL). Unexpectedly, nAChR antagonism not only abolished nicotine-induced HMVEC migration but also abolished migration induced by bFGF and attenuated migration induced by VEGF. Transcriptional profiling identified gene expression programs which were concordantly regulated by all 3 angiogens (nicotine, VEGF, and bFGF), a notable feature of which includes corepression of thioredoxin-interacting protein (TXNIP), endogenous inhibitor of the redox regulator thioredoxin. Furthermore, TXNIP repression by all 3 angiogens induced thioredoxin activity. Silencing thioredoxin by small interference RNA abrogated all angiogen-induced migration while silencing TXNIP strongly induced HMVEC migration. Interestingly, nAChR antagonism abrogates growth factor (VEGF and bFGF)-mediated induction of thioredoxin activity. CONCLUSIONS Nicotine promotes angiogenesis via stimulation of nAChR-dependent endothelial cell migration. Furthermore, growth factor-induced HMVEC migration, a key angiogenesis event, requires nAChR activation--an effect mediated in part by nAChR-dependent regulation of thioredoxin activity.
Collapse
MESH Headings
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cell Movement/drug effects
- Cell Movement/physiology
- Cells, Cultured
- Dose-Response Relationship, Drug
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/physiology
- Fibroblast Growth Factor 2/physiology
- Gene Expression Regulation/drug effects
- Humans
- Neovascularization, Physiologic/drug effects
- Neovascularization, Physiologic/physiology
- Nicotine/pharmacology
- Nicotinic Agonists/pharmacology
- RNA, Small Interfering/pharmacology
- Receptors, Nicotinic/genetics
- Receptors, Nicotinic/physiology
- Thioredoxins/genetics
- Thioredoxins/metabolism
- Vascular Endothelial Growth Factor A/physiology
Collapse
Affiliation(s)
- Martin K C Ng
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Barcaroli D, Dinsdale D, Neale MH, Bongiorno-Borbone L, Ranalli M, Munarriz E, Sayan AE, McWilliam JM, Smith TM, Fava E, Knight RA, Melino G, De Laurenzi V. FLASH is an essential component of Cajal bodies. Proc Natl Acad Sci U S A 2006; 103:14802-7. [PMID: 17003126 PMCID: PMC1578500 DOI: 10.1073/pnas.0604225103] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cajal bodies are small nuclear organelles with a number of nuclear functions. Here we show that FLICE-associated huge protein (FLASH), originally described as a component of the apoptosis signaling pathway, is mainly localized in Cajal bodies and is essential for their structure. Reduction in FLASH expression by short hairpin RNA results in disruption of the normal architecture of the Cajal body and relocalization of its components. Because the function of FLASH in the apoptosis receptor signaling pathway has been strongly questioned, we have now identified a clear function for this protein.
Collapse
Affiliation(s)
- D. Barcaroli
- *Medical Research Council, Toxicology Unit, Leicester University, Hodgkin Building, Lancaster Road, P.O. Box 138, Leicester LE1 9HN, United Kingdom
- Fondazione S. Lucia, Istituto di Ricovero e Cura a Carattere Scientifico, Centro Europeo di Ricerca sul Cervello, Via del Fosso di Fiorano 64/65, 00143 Rome, Italy
| | - D. Dinsdale
- *Medical Research Council, Toxicology Unit, Leicester University, Hodgkin Building, Lancaster Road, P.O. Box 138, Leicester LE1 9HN, United Kingdom
| | - M. H. Neale
- *Medical Research Council, Toxicology Unit, Leicester University, Hodgkin Building, Lancaster Road, P.O. Box 138, Leicester LE1 9HN, United Kingdom
| | - L. Bongiorno-Borbone
- Department of Experimental Medicine, University of Rome “Tor Vergata,” Via Montpellier 1, 00133 Rome, Italy; and
- Fondazione S. Lucia, Istituto di Ricovero e Cura a Carattere Scientifico, Centro Europeo di Ricerca sul Cervello, Via del Fosso di Fiorano 64/65, 00143 Rome, Italy
| | - M. Ranalli
- Department of Experimental Medicine, University of Rome “Tor Vergata,” Via Montpellier 1, 00133 Rome, Italy; and
| | - E. Munarriz
- *Medical Research Council, Toxicology Unit, Leicester University, Hodgkin Building, Lancaster Road, P.O. Box 138, Leicester LE1 9HN, United Kingdom
| | - A. E. Sayan
- *Medical Research Council, Toxicology Unit, Leicester University, Hodgkin Building, Lancaster Road, P.O. Box 138, Leicester LE1 9HN, United Kingdom
| | - J. M. McWilliam
- *Medical Research Council, Toxicology Unit, Leicester University, Hodgkin Building, Lancaster Road, P.O. Box 138, Leicester LE1 9HN, United Kingdom
| | - T. M. Smith
- *Medical Research Council, Toxicology Unit, Leicester University, Hodgkin Building, Lancaster Road, P.O. Box 138, Leicester LE1 9HN, United Kingdom
| | - E. Fava
- *Medical Research Council, Toxicology Unit, Leicester University, Hodgkin Building, Lancaster Road, P.O. Box 138, Leicester LE1 9HN, United Kingdom
| | - R. A. Knight
- *Medical Research Council, Toxicology Unit, Leicester University, Hodgkin Building, Lancaster Road, P.O. Box 138, Leicester LE1 9HN, United Kingdom
| | - G. Melino
- *Medical Research Council, Toxicology Unit, Leicester University, Hodgkin Building, Lancaster Road, P.O. Box 138, Leicester LE1 9HN, United Kingdom
- Department of Experimental Medicine, University of Rome “Tor Vergata,” Via Montpellier 1, 00133 Rome, Italy; and
- Fondazione S. Lucia, Istituto di Ricovero e Cura a Carattere Scientifico, Centro Europeo di Ricerca sul Cervello, Via del Fosso di Fiorano 64/65, 00143 Rome, Italy
| | - V. De Laurenzi
- *Medical Research Council, Toxicology Unit, Leicester University, Hodgkin Building, Lancaster Road, P.O. Box 138, Leicester LE1 9HN, United Kingdom
- Department of Experimental Medicine, University of Rome “Tor Vergata,” Via Montpellier 1, 00133 Rome, Italy; and
- Fondazione S. Lucia, Istituto di Ricovero e Cura a Carattere Scientifico, Centro Europeo di Ricerca sul Cervello, Via del Fosso di Fiorano 64/65, 00143 Rome, Italy
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
21
|
Abstract
AbstractAngiotensin-converting enzyme (ACE, kininase II) is a plasma membrane zinc metallopeptidase that acts as a key enzyme for the extracellular conversion of vasoactive peptides. Recently, ACE outside-in signalling in endothelial cells has been described. The present study tested the hypothesis that ACE signalling is not restricted to endothelial cells and may act as an additional peptide receptor on human preadipocytes and adipocytes. ACE protein levels were not changed during adipose conversion of human primary preadipocytes. The enzyme was primarily localized to the non-detergent-resistant fraction of the membrane and phosphorylated in non-dividing cells. Antibody arrays of whole cell lysate detected putative ACE-interacting proteins, which all share important roles in cell cycle control and/or apoptosis. These findings suggest that ACE is a versatile molecule, involved both in the regulation of extracellular peptide concentrations and direct intracellular signalling. In human adipose cells ACE may potentially influence exit from the cell cycle, differentiation, and programmed cell death signalling.
Collapse
|
22
|
Park SG, Ryu HM, Lim SO, Kim YI, Hwang SB, Jung G. Interferon-gamma inhibits hepatitis B virus-induced NF-kappaB activation through nuclear localization of NF-kappaB-inducing kinase. Gastroenterology 2005; 128:2042-53. [PMID: 15940636 DOI: 10.1053/j.gastro.2005.03.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Nuclear factor-kappaB (NF-kappaB) signaling pathway is an important regulating pathway in liver diseases, including hepatocellular carcinoma. In our study, immunohistochemical analysis showed that NF-kappaB-inducing kinase (NIK), an upstream kinase of IkappaB kinases, nuclear localization occurs only in liver tissues obtained from hepatitis B surface antigen (HBsAg)(+) patients but not in tissues from HBsAg(-) patients. The aim of the present study was to identify the inducer of NIK nuclear localization and determine whether the NIK nuclear localization affects the hepatitis B virus (HBV)-mediated NF-kappaB activation. METHODS The experiments were performed on HepG2.2.15 cells and on HepG2 cells transfected with pHBV1.2x, a plasmid encoding all HBV messages, using NF-kappaB-dependent luciferase reporter gene assay, electrophoretic mobility shift assay, immunoblot analysis, and fluorescent microscopy analysis. RESULTS HBV induced NIK-dependent NF-kappaB activation. However, interferon (IFN)-gamma induced NIK nuclear localization and inhibited NF-kappaB activation in HepG2.2.15 cells and in HepG2 cells transfected with pHBV1.2x. When NIK nuclear localization was inhibited by deletion of nuclear localization signal on NIK, IFN-gamma did not induce the NIK nuclear localization and did not inhibit NF-kappaB activation. CONCLUSIONS IFN-gamma selectively inhibits HBV-mediated NF-kappaB activation. This inhibition is accomplished by NIK nuclear localization, which is a novel mechanism of NF-kappaB inhibition.
Collapse
Affiliation(s)
- Sung Gyoo Park
- School of Biological Sciences, Seoul National University, Korea
| | | | | | | | | | | |
Collapse
|
23
|
Pyo JO, Jang MH, Kwon YK, Lee HJ, Jun JI, Woo HN, Cho DH, Choi B, Lee H, Kim JH, Mizushima N, Oshumi Y, Jung YK. Essential roles of Atg5 and FADD in autophagic cell death: dissection of autophagic cell death into vacuole formation and cell death. J Biol Chem 2005; 280:20722-9. [PMID: 15778222 DOI: 10.1074/jbc.m413934200] [Citation(s) in RCA: 423] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Autophagic cell death is characterized by the accumulation of vacuoles in physiological and pathological conditions. However, its molecular event is unknown. Here, we show that Atg5, which is known to function in autophagy, contributes to autophagic cell death by interacting with Fas-associated protein with death domain (FADD). Down-regulation of Atg5 expression in HeLa cells suppresses cell death and vacuole formation induced by IFN-gamma. Inversely, ectopic expression of Atg5 using adenoviral delivery induces autophagic cell death. Deletion mapping analysis indicates that procell death activity resides in the middle and C-terminal region of Atg5. Cells harboring the accumulated vacuoles triggered by IFN-gamma or Atg5 expression become dead, and vacuole formation precedes cell death. 3-Methyladenine or expression of Atg5(K130R) mutant blocks both cell death and vacuole formation triggered by IFN-gamma, whereas benzyloxycarbonyl-VAD-fluoromethyl ketone (Z-VAD-fmk) inhibits only cell death but not vacuole formation. Atg5 interacts with FADD via death domain in vitro and in vivo, and the Atg5-mediated cell death, but not vacuole formation, is blocked in FADD-deficient cells. These results suggest that Atg5 plays a crucial role in IFN-gamma-induced autophagic cell death by interacting with FADD.
Collapse
Affiliation(s)
- Jong-Ok Pyo
- Department of Life Science, Gwangju Institute of Science and Technology, Gwangju 500-712, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Jun JI, Chung CW, Lee HJ, Pyo JO, Lee KN, Kim NS, Kim YS, Yoo HS, Lee TH, Kim E, Jung YK. Role of FLASH in caspase-8-mediated activation of NF-kappaB: dominant-negative function of FLASH mutant in NF-kappaB signaling pathway. Oncogene 2005; 24:688-96. [PMID: 15592525 DOI: 10.1038/sj.onc.1208186] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Caspase-8 is the most receptor-proximal, upstream caspase in the caspase cascade and plays a key role in cell death triggered by various death receptors. Here, we addressed the role of endogenous caspase-8 in tumor necrosis factor (TNF)-alpha-induced activation of NF-kappaB. Direct targeting of caspase-8 with siRNA and antisense (AS) approaches abolished TNF-alpha-induced activation of NF-kappaB in NIH3T3, HeLa, and HEK293 cells as determined with luciferase reporter gene and cell fractionation assays. Reconstitution of caspase-8-deficient C33A cells with processing-defective (P/D) mutant of caspase-8 sensitized the cells to TNF-alpha for NF-kappaB activation. In contrast to wild-type caspase-8, death effector domain mutant replacing Asp73 with Ala (caspase-8 (D73A)) failed to activate NF-kappaB and to bind FLICE-associated huge protein (FLASH) in vitro and in vivo. Instead, caspase-8 (D73A) mutant bound to caspase-8 and blocked NF-kappaB activation triggered by TNF-alpha and caspase-8. In addition, expression of an NF-kappaB-activating domain-deletion mutant of FLASH or transfection of FLASH AS oligonucleotides abolished TNF-alpha and caspase-8, but not phorbol 12-myristate 13-acetate, -induced activation of NF-kappaB. Further, immunoprecipitation assays showed that caspase-8 formed triple complex with TRAF2 and FLASH. Taken together, these results suggest that endogenous caspase-8 mediates TNF-alpha-induced activation of NF-kappaB via FLASH.
Collapse
Affiliation(s)
- Joon-Il Jun
- Department of Life Science, Gwangju Institute of Science and Technology, 1-Oryong-dong, Buk-gu, Gwangju 500-712, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Woo HN, Hong GS, Jun JI, Cho DH, Choi HW, Lee HJ, Chung CW, Kim IK, Jo DG, Pyo JO, Bertin J, Jung YK. Inhibition of Bcl10-mediated activation of NF-kappa B by BinCARD, a Bcl10-interacting CARD protein. FEBS Lett 2005; 578:239-44. [PMID: 15637807 DOI: 10.1016/j.febslet.2004.10.094] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
We have identified a novel CARD-containing protein from EST database. BinCARD (Bcl10-interacting protein with CARD). BinCARD was ubiquitously expressed. Co-immunoprecipitation, In vitro binding, mammalian two-hybrid, and immunostaining assays revealed that BinCARD interacted with Bcl10 through CARD. BinCARD potently suppressed NF-kappa B activation induced by Bcl10 and decreased the amounts of phosphorylated Bcl10. Mutations at the residue Leu17 or Leu65, which is highly conserved in CARD, abolished the inhibitory effects of BinCARD on both Bcl10-induced activation of NF-kappa B and phosphorylation of Bcl10. Further, expression of BinCARD inhibited Bcl10 phosphorylation induced by T cell activation signal. These results suggest that BinCARD interacts with Bcl10 to inhibit Bcl10-mediated activation of NF-kappa B and to suppress Bcl10 phosphorylation.
Collapse
Affiliation(s)
- Ha-Na Woo
- Department of Life Science, Gwangju Institute of Science and Technology, Gwangju 500-712, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
The developing limb bud provides one of the best examples in which programmed cell death exerts major morphogenetic functions. In this work, we revise the distribution and the developmental significance of cell death in the embryonic vertebrate limb and its control by the BMP signalling pathway. In addition, paying special attention to the interdigital apoptotic zones, we review current data concerning the intracellular death machinery implicated in mesodermal limb apoptosis.
Collapse
Affiliation(s)
- Vanessa Zuzarte-Luis
- Departamento de Anatomia y Biologia Celular, Universidad de Cantabria, C/Cardenal Herrera Oria, s/n, 39011 Santander, Cantabria, Spain
| | | |
Collapse
|
27
|
Niculescu MD, Craciunescu CN, Zeisel SH. Gene expression profiling of choline-deprived neural precursor cells isolated from mouse brain. ACTA ACUST UNITED AC 2004; 134:309-22. [PMID: 15836926 DOI: 10.1016/j.molbrainres.2004.11.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2004] [Revised: 11/01/2004] [Accepted: 11/04/2004] [Indexed: 12/16/2022]
Abstract
Choline is an essential nutrient and an important methyl donor. Choline deficiency alters fetal development of the hippocampus in rodents and these changes are associated with decreased memory function lasting throughout life. Also, choline deficiency alters global and gene-specific DNA methylation in several models. This gene expression profiling study describes changes in cortical neural precursor cells from embryonic day 14 mice, after 48 h of exposure to a choline-deficient medium. Using Significance Analysis of Microarrays, we found the expression of 1003 genes to be significantly changed (from a total of 16,000 total genes spotted on the array), with a false discovery rate below 5%. A total of 846 genes were overexpressed while 157 were underexpressed. Classification by gene ontology revealed that 331 of these genes modulate cell proliferation, apoptosis, neuronal and glial differentiation, methyl metabolism, and calcium-binding protein classes. Twenty-seven genes that had changed expression have previously been reported to be regulated by promoter or intron methylation. These findings support our previous work suggesting that choline deficiency decreases the proliferation of neural precursors and possibly increases premature neuronal differentiation and apoptosis.
Collapse
Affiliation(s)
- Mihai D Niculescu
- Department of Nutrition, School of Public Health and School of Medicine, University of North Carolina at Chapel Hill, 2212 McGavran-Greenberg Hall, Chapel Hill, NC 27599-7461, USA
| | | | | |
Collapse
|
28
|
Kino T, Ichijo T, Chrousos GP. FLASH interacts with p160 coactivator subtypes and differentially suppresses transcriptional activity of steroid hormone receptors. J Steroid Biochem Mol Biol 2004; 92:357-63. [PMID: 15698540 DOI: 10.1016/j.jsbmb.2004.09.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2003] [Accepted: 09/09/2004] [Indexed: 11/30/2022]
Abstract
We previously reported that tumor necrosis factor alpha receptor- and Fas-associated FLASH interacts with one of the p160 nuclear receptor coactivators, glucocorticoid receptor-interacting protein (GRIP) 1, at its nuclear receptor-binding (NRB) domain, and that inhibits the transcriptional activity of the glucocorticoid receptor (GR) by interfering with association of GR and GRIP1. Here, we further examined the specificity of FLASH suppressive effect and the physical/functional interactions between this protein and two other p160 family subtypes. The suppressive effect of FLASH on GR transactivation was observed in several cell lines and on the chromatin-integrated mouse mammary tumor virus (MMTV) promoter. FLASH strongly interacted with the NRB domain of the thyroid hormone receptor activator molecule (TRAM) 1, a member of the steroid hormone receptor coactivator (SRC) 3/nuclear receptor coactivator (N-CoA) 3 subtypes, as well as with SRC2/N-CoA2 p160 coactivator GRIP1, while its interaction with SRC1a, one of the SRC1/N-CoA1 proteins, was faint in yeast two-hybrid assays. Accordingly, FLASH strongly suppressed TRAM1- and GRIP1-induced enhancement of GR-stimulated transactivation of the MMTV promoter in HCT116 cells, while it did not affect SRC1a-induced potentiation of transcription. Furthermore, FLASH suppressed androgen- and progesterone receptor-induced transcriptional activity, but did not influence estrogen receptor-induced transactivation, possibly due to their preferential use of p160 coactivators in HCT116 and HeLa cells. Thus, FLASH differentially suppresses steroid hormone receptor-induced transcriptional activity by interfering with their association with SRC2/N-CoA2 and SRC3/N-CoA3 but not with SRC1/N-CoA1.
Collapse
Affiliation(s)
- Tomoshige Kino
- Pediatric and Reproductive Endocrinology Branch, National Institute of Child Health and Human Development, Building 10, Clinical Research Center, Room 1-3140 , 10 Center Drive, MSC 1109, Bethesda, MD 20892-1109, USA.
| | | | | |
Collapse
|
29
|
Lee SH, Park SG, Lim SO, Jung G. The hepatitis B virus X protein up-regulates lymphotoxin alpha expression in hepatocytes. Biochim Biophys Acta Mol Basis Dis 2004; 1741:75-84. [PMID: 15955450 DOI: 10.1016/j.bbadis.2004.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2004] [Revised: 09/25/2004] [Accepted: 10/05/2004] [Indexed: 10/26/2022]
Abstract
Hepatitis B virus X protein (HBx) is involved in intrahepatic inflammatory processes by inducing several pro-inflammatory cytokines. It has been suggested that these inflammatory processes play an important role in causing hepatocarcinogenesis. In this study, we investigated the role of HBx in the expression of lymphotoxin alpha (LTalpha) in hepatoma cells such as Huh-7 and Chang. Our experiments showed that (i) transient HBx expression in Huh-7 cells up-regulated LTalpha mRNA expression; (ii) this up-regulation was predominantly affected by HBx-induced nuclear factor-kappaB (NF-kappaB) activation. In addition, the HBx-induced NF-kappaB activation was decreased by the treatment of LTalpha neutralizing antibodies in a dose-dependent manner. We conclude that HBx up-regulates LTalpha expression at the transcriptional level through an NF-kappaB-dependent mechanism and, therefore, the up-regulated LTalpha may be secreted and involved in the HBx-induced NF-kappaB activation.
Collapse
Affiliation(s)
- Sang Hun Lee
- School of Biological Sciences, Seoul National University, Seoul 151-747, Republic of Korea
| | | | | | | |
Collapse
|
30
|
Konstantinov IE, Arab S, Kharbanda RK, Li J, Cheung MMH, Cherepanov V, Downey GP, Liu PP, Cukerman E, Coles JG, Redington AN. The remote ischemic preconditioning stimulus modifies inflammatory gene expression in humans. Physiol Genomics 2004; 19:143-50. [PMID: 15304621 DOI: 10.1152/physiolgenomics.00046.2004] [Citation(s) in RCA: 257] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Remote ischemic preconditioning (IPC) reduces tissue injury caused by ischemia-reperfusion (IR) in distant organs. We tested the hypothesis that remote IPC (rIPC) modifies inflammatory gene transcription in humans. Using a microarray method, we demonstrated that a simple model of brief forearm ischemia suppresses proinflammatory gene expression in circulating leukocytes. Genes encoding key proteins involved in cytokine synthesis, leukocyte chemotaxis, adhesion and migration, exocytosis, innate immunity signaling pathways, and apoptosis were all suppressed within 15 min (early phase IPC) and more so after 24 h (second window IPC). Changes in leukocyte CD11b expression measured by flow cytometry mirrored this pattern, with there being a significant (P = 0.01) reduction at 24 h. The results of this study show that the rIPC stimulus modifies leukocyte inflammatory gene expression. This effect may contribute to the protective effect of IPC against IR injury and may have broader implications in other inflammatory processes. This is the first study of human gene expression following rIPC stimulus. rIPC stimulus suppressed proinflammatory gene transcription in human leukocytes.
Collapse
Affiliation(s)
- Igor E Konstantinov
- Division of Cardiovascular Surgery, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Morgan R, Nalliah A, Morsi El-Kadi AS. FLASH, a component of the FAS-CAPSASE8 apoptotic pathway, is directly regulated by Hoxb4 in the notochord. Dev Biol 2004; 265:105-12. [PMID: 14697356 DOI: 10.1016/j.ydbio.2003.09.030] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The Hox genes are a family of homeodomain-containing transcription factors that confer positional identity during development. Although their regulation and function have been extensively studied, very little is known of their downstream target genes. Here we show that Hoxb4 directly induces the expression of FLASH in the notochord of embryos after neurulation. FLASH is a component of the FAS-CAPSASE8 apoptotic pathway, and blocking its activity, or that of Hoxb4, prevents apoptosis in the notochord.
Collapse
Affiliation(s)
- Richard Morgan
- Department of Anatomy and Developmental Biology, St. George's Hospital Medical School, Cranmer Terrace, London SW17 0RE, UK.
| | | | | |
Collapse
|
32
|
Abstract
Apoptosis, also known as programmed cell death, is the major type of cell death involved in normal development, regeneration, proliferation and pathologic degeneration in the central nervous system (CNS). The apoptotic process can be divided further into two pathways depending on the involvement of mitochondria and related biochemical cascades. The internal pathway of apoptosis is initiated by a variety of cytotoxic stimuli and mediated by the release of cytochrome c and subsequent activation of downstream caspases. The external pathway is mainly triggered by ligation of death receptors such as Fas, tumor necrosis factor (TNF)-related apoptosis inducing ligand-R1 (TRAIL-R1), TRAIL-R2 and TNFRp55, and mediated by direct activation of upstream caspases. The Fas-FasL system has been known as a prototypic inducer of extrinsic cell death responsible for cell-mediated cytotoxicity, peripheral immune regulation, immune privilege and "counterattack" of malignant tumor cells against the host immune system. Fas and FasL are expressed in the normal CNS, and expression increases in inflamed and degenerated brains. Like other specialized tissues such as the eye and testis, the Fas-FasL system is thought to be involved in immune suppressed status in the CNS. Expression of Fas and FasL is significantly elevated in a variety of the neurologic disorders, suggesting the possibility that this system may play roles in degenerative and inflammatory responses in the CNS. Therefore, the FasL-Fas system should be considered as a double-edged sword in the CNS: maintaining the immune suppressed status in normal brain and inducing neuronal cell death and inflammation in a variety of neurologic disorders.
Collapse
Affiliation(s)
- Chulhee Choi
- The Center for Cell Signaling Research and Division of Molecular Life Sciences, Ewha Womans University, 11-1 Daehyun-dong, Sudaemun-gu, Seoul 120-750, South Korea.
| | | |
Collapse
|
33
|
Abstract
Apoptosis signaling is regulated and executed by specialized proteins that often carry protein/protein interaction domains. One of these domains is the death effector domain (DED) that is predominantly found in components of the death-inducing signaling complex, which forms at the members of the death receptor family following their ligation. Both proapoptotic- and antiapoptotic-DED-containing proteins have been identified, which makes these proteins exquisitely suited to the regulation of apoptosis. Aside from their pivotal role in the control of the apoptotic program, DED-containing proteins have recently been demonstrated to exert their influence on other cellular processes as well, including cell proliferation. These data highlight the multiple roles for the members of this family, suggesting that they are suited to control both life and death decisions of cells. Additionally, because they can act proapoptotically, antiapoptotically, or in the regulation of the cell cycle, this family of proteins may be excellent candidates for cancer therapy targets. Oncogene (2003) 22, 8634-8644. doi:10.1038/sj.onc.1207103
Collapse
Affiliation(s)
- Bryan C Barnhart
- The Ben May Institute for Cancer Research, University of Chicago, 924 E 57th Street, Chicago, IL 60637, USA
| | | | | | | |
Collapse
|
34
|
Park MY, Jang HD, Lee SY, Lee KJ, Kim E. Fas-associated factor-1 inhibits nuclear factor-kappaB (NF-kappaB) activity by interfering with nuclear translocation of the RelA (p65) subunit of NF-kappaB. J Biol Chem 2003; 279:2544-9. [PMID: 14600157 DOI: 10.1074/jbc.m304565200] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Fas-associated factor-1 (FAF1) is a Fas-binding pro-apoptotic protein that is a component of the death-inducing signaling complex in Fas-mediated apoptosis. Here, we show that FAF1 is involved in negative regulation of NF-kappaB activation. Overexpression of FAF1 decreased the basal level of NF-kappaB activity in 293 cells. NF-kappaB activation induced by tumor necrosis factor (TNF)-alpha, interleukin-1beta, and lipopolysaccharide was also inhibited by FAF1 overexpression. Moreover, FAF1 suppressed NF-kappaB activation induced by transducers of diverse NF-kappaB-activating signals such as TNF receptor-associated factor-2 and -6, MEKK1, and IkappaB kinase-beta as well as NF-kappaB p65, one of the end point molecules in the NF-kappaB activation pathway, suggesting that NF-kappaB p65 might be a target molecule upon which FAF1 acts. Subsequent study disclosed that FAF1 physically interacts with NF-kappaB p65 and that the binding domain of FAF1 is the death effector domain (DED)-interacting domain (amino acids 181-381), where DEDs of the Fas-associated death domain protein and caspase-8 interact. The NF-kappaB activity-modulating potential of FAF1 was also mapped to the DED-interacting domain. Finally, overexpression of FAF1 prevented translocation of NF-kappaB p65 into the nucleus and decreased its DNA-binding activity upon TNFalpha treatment. This study presents a novel function of FAF1, in addition to the previously known function as a component of the Fas death-inducing signaling complex, i.e. NF-kappaB activity suppressor by cytoplasmic retention of NF-kappaB p65 via physical interaction.
Collapse
Affiliation(s)
- Min-Young Park
- Research Center for Biomedicinal Resources and the Division of Life Science, PaiChai University, Daejeon 302-735, Korea
| | | | | | | | | |
Collapse
|
35
|
Kino T, De Martino MU, Charmandari E, Mirani M, Chrousos GP. Tissue glucocorticoid resistance/hypersensitivity syndromes. J Steroid Biochem Mol Biol 2003; 85:457-67. [PMID: 12943736 DOI: 10.1016/s0960-0760(03)00218-8] [Citation(s) in RCA: 119] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Glucocorticoids have a broad array of life-sustaining functions and play an important role in the therapy of many diseases. Thus, changes of tissue sensitivity to glucocorticoids may be associated with and influence the course and treatment of many pathologic states. Such tissue sensitivity changes may present on either side of an optimal range, respectively as glucocorticoid resistance or hypersensitivity, and may be generalized or tissue-specific. Familial/sporadic glucocorticoid resistance syndrome caused by inactivating mutations of the glucocorticoid receptor (GR) gene is a classic monogenic disorder associated with congenital, generalized glucocorticoid insensitivity, while several autoimmune, inflammatory and allergic diseases are often associated with resistance of the inflamed tissues to glucocorticoids. On the other hand, glucocorticoid hypersensitivity has been suggested in visceral obesity-related insulin resistance associated with components of the metabolic syndrome, and in the acquired immunodeficiency syndrome (AIDS) caused by human immunodeficiency virus type-1 (HIV-1) infection. Here, we have reviewed the molecular analyses of five familial and three sporadic cases of the familial/sporadic glucocorticoid resistance syndrome and discussed the possible contribution of newly identified molecules, such as HIV-1 accessory proteins Vpr and Tat, FLICE-associated huge protein (FLASH) and chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII), on the molecular regulation of GR activity, as well as their possible contribution to changes in tissue sensitivity to glucocorticoids in pathologic conditions.
Collapse
Affiliation(s)
- Tomoshige Kino
- Pediatric and Reproductive Endocrinology Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1583, USA.
| | | | | | | | | |
Collapse
|
36
|
Kino T, Chrousos GP. Tumor necrosis factor alpha receptor- and Fas-associated FLASH inhibit transcriptional activity of the glucocorticoid receptor by binding to and interfering with its interaction with p160 type nuclear receptor coactivators. J Biol Chem 2003; 278:3023-9. [PMID: 12477726 DOI: 10.1074/jbc.m209234200] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Tumor necrosis factor alpha (TNF alpha) and its downstream transcription factor nuclear factor kappa B (NF-kappa B) suppress glucocorticoid action, contributing to tissue resistance to glucocorticoids in several pathologic inflammatory states. p160 nuclear receptor coactivators on the other hand, contribute to the transcriptional signal of the glucocorticoid receptor (GR) through interaction with it via LXXLL motifs in their nuclear receptor-binding (NRB) domain. To discover TNF alpha-induced factors that regulate GR activity at the coactivator level, we performed yeast two-hybrid screening using the NRB domain of the glucocorticoid receptor-interacting protein 1 (GRIP1) as bait. We found that FLICE-associated huge protein (FLASH), which transduces TNF alpha and Fas ligand signals, bound the NRB domain of GRIP1 at a region between the second and third LXXLL motifs. FLASH suppressed both GR transactivation and GRIP1 enhancement of the glucocorticoid signal and inhibited the physical interaction between GR and the GRIP1 NRB domain. Transfected green fluorescent protein-fused FLASH was located in both the cytoplasm and nucleus, while endogenous FLASH shifted its subcellular localization from the cytoplasm into the nucleus in response to TNF alpha. FLASH antisense and super-repressor I kappa B alpha inhibited the action of TNF alpha independently of each other and additively. These findings indicate that FLASH participates in TNF alpha-induced blockade of GR transactivation at the nuclear receptor coactivator level, upstream and independently of NF-kappa B.
Collapse
Affiliation(s)
- Tomoshige Kino
- Pediatric and Reproductive Endocrinology Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892-1583, USA.
| | | |
Collapse
|
37
|
Arron JR, Walsh MC, Choi Y. TRAF‐MediatedTNFR‐Family Signaling. ACTA ACUST UNITED AC 2002; Chapter 11:11.9D.1-11.9D.14. [DOI: 10.1002/0471142735.im1109ds51] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Joseph R. Arron
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine Philadelphia Pennsylvania
| | - Matthew C. Walsh
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine Philadelphia Pennsylvania
| | - Yongwon Choi
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine Philadelphia Pennsylvania
| |
Collapse
|
38
|
Bremner P, Heinrich M. Natural products as targeted modulators of the nuclear factor-kappaB pathway. J Pharm Pharmacol 2002; 54:453-72. [PMID: 11999122 DOI: 10.1211/0022357021778637] [Citation(s) in RCA: 218] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The use of plant extracts to alleviate inflammatory diseases is centuries old and continues to this day. This review assesses the current understanding of the use of such plants and natural products isolated from them in terms of their action against the ubiquitous transcription factor, nuclear factor kappa B (NF-kappaB). As an activator of many pro-inflammatory cytokines and inflammatory processes the modulation of the NF-kappaB transduction pathway is a principal target to alleviate the symptoms of such diseases as arthritis, inflammatory bowel disease and asthma. Two pathways of NF-kappaB activation will first be summarised, leading to the IKK (IkappaB kinase) complex, that subsequently initiates phosphorylation of the NF-kappaB inhibitory protein (IKB). Natural products and some extracts are reviewed and assessed for their activity and potency as NF-kappaB inhibitors. A large number of compounds are currently known as NF-kappaB modulators and include the isoprenoids, most notably kaurene diterpenoids and members of the sesquiterpene lactones class, several phenolics including curcumin and flavonoids such as silybin. Additional data on cellular toxicity are also highlighted as an exclusion principle for pursuing such compounds in clinical development. In addition, where enough data exists some conclusions on structure-activity relationship are provided.
Collapse
Affiliation(s)
- Paul Bremner
- Centre for Pharmacognosy and Phytotherapy, School of Pharmacy, London, UK.
| | | |
Collapse
|
39
|
Lyon GD, Newton AC, Marshall B. The need for a standard nomenclature for gene classification (a Nucleotide Function Code) and an automated data-based tool to assist in understanding the molecular associations in cell signalling in plant-pathogen interactions. MOLECULAR PLANT PATHOLOGY 2002; 3:103-109. [PMID: 20569315 DOI: 10.1046/j.1464-6722.2002.00101.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
summary Despite the adoption of Arabidopsis thaliana as a model plant system and the plethora of molecular information being obtained from its use, it is disappointing that the scientific community has not devised a cell signalling model integrating and visualizing these data. Lack of common systems of nomenclature and the sheer size and complexity of the task inhibit any individual from bringing together the knowledge into a unified structure. There are clearly many aspects of cell biology that are similar, even between plants and animals, that could facilitate development of a generic model. A gene-coding or nucleotide classification system which is 'user-friendly' would be beneficial to building such a model and enable rapid identification of orthologues of genes from different organisms. Whilst some international projects seek to address the problem of assigning unique numbers to genes, none suggest a nucleotide classification system that provides biological information that is transparent within the code. This paper discusses these issues and identifies the need for a more formal, semi-automated approach to modelling signal transduction utilizing the strengths of the proposed classification approach. By way of illustration, an example of a possible nucleotide function code is suggested, to demonstrate more clearly the benefits of such a system. Further discussion of this topic will be encouraged on websites (<www.bspp.org.uk> and <www.drastic.org.uk>).
Collapse
Affiliation(s)
- Gary D Lyon
- Scottish Crop Research Institute, Invergowrie, Dundee DD2 5DA, Scotland, UK
| | | | | |
Collapse
|