51
|
Xiao Y, Motomura S, Deyev V, Podack ER. TNF superfamily member 13, APRIL, inhibits allergic lung inflammation. Eur J Immunol 2010; 41:164-71. [PMID: 21182087 DOI: 10.1002/eji.201040436] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Revised: 08/12/2010] [Accepted: 10/15/2010] [Indexed: 01/01/2023]
Abstract
The T-cell functions of a proliferation-inducing ligand (APRIL, also known as TNFSF13) remain largely undefined. We previously showed that APRIL suppressed Th2 cytokine production in cultured CD4(+) T cells and Th2 antibody responses. Here we show that APRIL suppresses allergic lung inflammation, which is associated with diminished expression of the transcription factor c-maf. Mice deficient in the April gene (April(-/-) mice) had significantly aggravated lung inflammation compared with WT mice in the ovalbumin-induced allergic lung inflammation model. Likewise, blockade of APRIL in WT mice by the APRIL-receptor fusion protein, transmembrane activator and calcium modulator and cyclophilin ligand interactor (TACI)-Ig, enhanced lung inflammation. Transfer of APRIL-sufficient, ovalbumin-specific, TCR-transgenic CD4(+) T (OT-II) cells to April(-/-) mice restored the suppressive effect of APRIL on lung inflammation. Mechanistically, the expression of the Th2 cytokine transcription factor c-maf, but not GATA-3, was markedly enhanced in April(-/-) CD4(+) T cells at the RNA and protein level and under non-polarizing (Th neutral, ThN) and Th2-polarizing conditions. Since c-maf transactivates the IL-4 gene, the increased c-maf expression in April(-/-) mice readily explains increased Th2 cytokine production. Independent of its effect on IL-4, APRIL suppressed IL-13 expression. APRIL thus may regulate lung inflammation in a dual way, by acting on c-maf expression and by directly controlling IL-13 production.
Collapse
Affiliation(s)
- Yanping Xiao
- Department of Microbiology & Immunology, University of Miami, Leonard Miller School of Medicine, Miami, FL, USA
| | | | | | | |
Collapse
|
52
|
Dalziel M, Kolesnichenko M, das Neves RP, Iborra F, Goding C, Furger A. Alpha-MSH regulates intergenic splicing of MC1R and TUBB3 in human melanocytes. Nucleic Acids Res 2010; 39:2378-92. [PMID: 21071418 PMCID: PMC3064779 DOI: 10.1093/nar/gkq1125] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Alternative splicing enables higher eukaryotes to increase their repertoire of proteins derived from a restricted number of genes. However, the possibility that functional diversity may also be augmented by splicing between adjacent genes has been largely neglected. Here, we show that the human melanocortin 1 receptor (MC1R) gene, a critical component of the facultative skin pigmentation system, has a highly complex and inefficient poly(A) site which is instrumental in allowing intergenic splicing between this locus and its immediate downstream neighbour tubulin-β-III (TUBB3). These transcripts, which produce two distinct protein isoforms localizing to the plasma membrane and the endoplasmic reticulum, seem to be restricted to humans as no detectable chimeric mRNA could be found in MC1R expressing mouse melanocytes. Significantly, treatment with the MC1R agonist α-MSH or activation of the stress response kinase p38-MAPK, both key molecules associated with ultraviolet radiation dermal insult and subsequent skin tanning, result in a shift in expression from MC1R in favour of chimeric MC1R-TUBB3 isoforms in cultured melanocytes. We propose that these chimeric proteins serve to equip melanocytes with novel cellular phenotypes required as part of the pigmentation response.
Collapse
Affiliation(s)
- Martin Dalziel
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | | | | | | | | | | |
Collapse
|
53
|
Lee SM, Kim WJ, Suk K, Lee WH. Cell to Cell Interaction Can Activate Membrane-bound APRIL Which Are Expressed on Inflammatory Macrophages. Immune Netw 2010; 10:173-80. [PMID: 21165246 PMCID: PMC2993949 DOI: 10.4110/in.2010.10.5.173] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Revised: 10/04/2010] [Accepted: 10/15/2010] [Indexed: 12/02/2022] Open
Abstract
Background APRIL, originally known as a cytokine involved in B cell survival, is now known to regulate the inflammatory activation of macrophages. Although the signal initiated from APRIL has been demonstrated, its role in cellular activation is still not clear due to the presence of BAFF, a closely related member of TNF superfamily, which share same receptors (TACI and BCMA) with APRIL. Methods Through transfection of siRNA, BAFF-deficient THP-1 cells (human macrophage-like cells) were generated and APRIL-mediated inflammatory activities were tested. The expression patterns of APRIL were also tested in vivo. Results BAFF-deficient THP-1 cells responded to APRIL-stimulating agents such as monoclonal antibody against APRIL and soluble form of TACI or BCMA. Furthermore, co-incubation of the siBAFF-deficient THP-1 cells with a human B cell line (Ramos) resulted in an activation of THP-1 cells which was dependent on interactions between APRIL and TACI/BCMA. Immunohistochemical analysis of human pathologic samples detected the expression of both APRIL and TACI in macrophage-rich areas. Additionally, human macrophage primary culture expressed APRIL on the cell surface. Conclusion These observations indicate that APRIL, which is expressed on macrophages in pathologic tissues with chronic inflammation, may mediate activation signals through its interaction with its counterparts via cell-to-cell interaction.
Collapse
Affiliation(s)
- Sang-Min Lee
- School of Life Sciences and Biotechnology, Kyungpook National University, Daegu 702-701, Korea
| | | | | | | |
Collapse
|
54
|
Prakash T, Sharma VK, Adati N, Ozawa R, Kumar N, Nishida Y, Fujikake T, Takeda T, Taylor TD. Expression of conjoined genes: another mechanism for gene regulation in eukaryotes. PLoS One 2010; 5:e13284. [PMID: 20967262 PMCID: PMC2953495 DOI: 10.1371/journal.pone.0013284] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Accepted: 09/14/2010] [Indexed: 11/19/2022] Open
Abstract
From the ENCODE project, it is realized that almost every base of the entire human genome is transcribed. One class of transcripts resulting from this arises from the conjoined gene, which is formed by combining the exons of two or more distinct (parent) genes lying on the same strand of a chromosome. Only a very limited number of such genes are known, and the definition and terminologies used for them are highly variable in the public databases. In this work, we have computationally identified and manually curated 751 conjoined genes (CGs) in the human genome that are supported by at least one mRNA or EST sequence available in the NCBI database. 353 representative CGs, of which 291 (82%) could be confirmed, were subjected to experimental validation using RT-PCR and sequencing methods. We speculate that these genes are arising out of novel functional requirements and are not merely artifacts of transcription, since more than 70% of them are conserved in other vertebrate genomes. The unique splicing patterns exhibited by CGs reveal their possible roles in protein evolution or gene regulation. Novel CGs, for which no transcript is available, could be identified in 80% of randomly selected potential CG forming regions, indicating that their formation is a routine process. Formation of CGs is not only limited to human, as we have also identified 270 CGs in mouse and 227 in drosophila using our approach. Additionally, we propose a novel mechanism for the formation of CGs. Finally, we developed a database, ConjoinG, which contains detailed information about all the CGs (800 in total) identified in the human genome. In summary, our findings reveal new insights about the functionality of CGs in terms of another possible mechanism for gene regulation and genomic evolution and the mechanism leading to their formation.
Collapse
Affiliation(s)
- Tulika Prakash
- MetaSystems Research Team, Computational Systems Biology Research Group, Advanced Computational Sciences Department, RIKEN Advanced Science Institute (ASI), Yokohama, Japan
| | - Vineet K. Sharma
- MetaSystems Research Team, Computational Systems Biology Research Group, Advanced Computational Sciences Department, RIKEN Advanced Science Institute (ASI), Yokohama, Japan
| | - Naoki Adati
- MetaSystems Research Team, Computational Systems Biology Research Group, Advanced Computational Sciences Department, RIKEN Advanced Science Institute (ASI), Yokohama, Japan
| | - Ritsuko Ozawa
- MetaSystems Research Team, Computational Systems Biology Research Group, Advanced Computational Sciences Department, RIKEN Advanced Science Institute (ASI), Yokohama, Japan
| | - Naveen Kumar
- MetaSystems Research Team, Computational Systems Biology Research Group, Advanced Computational Sciences Department, RIKEN Advanced Science Institute (ASI), Yokohama, Japan
| | - Yuichiro Nishida
- MetaSystems Research Team, Computational Systems Biology Research Group, Advanced Computational Sciences Department, RIKEN Advanced Science Institute (ASI), Yokohama, Japan
| | - Takayoshi Fujikake
- MetaSystems Research Team, Computational Systems Biology Research Group, Advanced Computational Sciences Department, RIKEN Advanced Science Institute (ASI), Yokohama, Japan
| | - Tadayuki Takeda
- MetaSystems Research Team, Computational Systems Biology Research Group, Advanced Computational Sciences Department, RIKEN Advanced Science Institute (ASI), Yokohama, Japan
| | - Todd D. Taylor
- MetaSystems Research Team, Computational Systems Biology Research Group, Advanced Computational Sciences Department, RIKEN Advanced Science Institute (ASI), Yokohama, Japan
- * E-mail:
| |
Collapse
|
55
|
Abstract
Ever since the pre-molecular era, the birth of new genes with novel functions has been considered to be a major contributor to adaptive evolutionary innovation. Here, I review the origin and evolution of new genes and their functions in eukaryotes, an area of research that has made rapid progress in the past decade thanks to the genomics revolution. Indeed, recent work has provided initial whole-genome views of the different types of new genes for a large number of different organisms. The array of mechanisms underlying the origin of new genes is compelling, extending way beyond the traditionally well-studied source of gene duplication. Thus, it was shown that novel genes also regularly arose from messenger RNAs of ancestral genes, protein-coding genes metamorphosed into new RNA genes, genomic parasites were co-opted as new genes, and that both protein and RNA genes were composed from scratch (i.e., from previously nonfunctional sequences). These mechanisms then also contributed to the formation of numerous novel chimeric gene structures. Detailed functional investigations uncovered different evolutionary pathways that led to the emergence of novel functions from these newly minted sequences and, with respect to animals, attributed a potentially important role to one specific tissue--the testis--in the process of gene birth. Remarkably, these studies also demonstrated that novel genes of the various types significantly impacted the evolution of cellular, physiological, morphological, behavioral, and reproductive phenotypic traits. Consequently, it is now firmly established that new genes have indeed been major contributors to the origin of adaptive evolutionary novelties.
Collapse
Affiliation(s)
- Henrik Kaessmann
- Center for Integrative Genomics, University of Lausanne, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
56
|
Brown SAN, Ghosh A, Winkles JA. Full-length, membrane-anchored TWEAK can function as a juxtacrine signaling molecule and activate the NF-kappaB pathway. J Biol Chem 2010; 285:17432-41. [PMID: 20385556 DOI: 10.1074/jbc.m110.131979] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor (TNF) family members are initially synthesized as type II transmembrane proteins, but some of these proteins are substrates for proteolytic enzymes that generate soluble cytokines with biological activity. TWEAK (TNF-like weak inducer of apoptosis), a member of the TNF family, is a multifunctional cytokine that acts via binding to a cell surface receptor named Fn14 (fibroblast growth factor-inducible 14). Studies conducted to date indicate that TWEAK-producing cells can co-express both membrane-anchored and soluble TWEAK isoforms, but there is little information on TWEAK proteolytic processing. Also, it is presently unclear whether membrane-anchored TWEAK, like soluble TWEAK, is biologically active. Here we show that full-length human TWEAK is processed intracellularly by the serine protease furin and identify TWEAK amino acid residues 90-93 as the predominant furin recognition site. In addition, we report that full-length, membrane-anchored TWEAK can bind the Fn14 receptor on neighboring cells and activate the NF-kappaB signaling pathway. Thus, TWEAK can act in a juxtacrine manner to initiate cellular responses, and this property may be important for TWEAK function during physiological wound repair and disease pathogenesis.
Collapse
Affiliation(s)
- Sharron A N Brown
- Department of Surgery and Physiology, Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | |
Collapse
|
57
|
Hegazy M, Darwish H, Darweesh H, El-Shehaby A, Emad Y. Raised serum level of APRIL in patients with systemic lupus erythematosus: Correlations with disease activity indices. Clin Immunol 2010; 135:118-24. [DOI: 10.1016/j.clim.2009.12.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Revised: 12/03/2009] [Accepted: 12/30/2009] [Indexed: 01/05/2023]
|
58
|
Dillon SR, Harder B, Lewis KB, Moore MD, Liu H, Bukowski TR, Hamacher NB, Lantry MM, Maurer M, Krejsa CM, Ellsworth JL, Pederson S, Elkon KB, Wener MH, Dall'Era M, Gross JA. B-lymphocyte stimulator/a proliferation-inducing ligand heterotrimers are elevated in the sera of patients with autoimmune disease and are neutralized by atacicept and B-cell maturation antigen-immunoglobulin. Arthritis Res Ther 2010; 12:R48. [PMID: 20302641 PMCID: PMC2888197 DOI: 10.1186/ar2959] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Revised: 02/12/2010] [Accepted: 03/19/2010] [Indexed: 12/14/2022] Open
Abstract
Introduction B-lymphocyte stimulator (BLyS) and a proliferation-inducing ligand (APRIL) are members of the tumor necrosis factor (TNF) family that regulate B-cell maturation, survival, and function. They are overexpressed in a variety of autoimmune diseases and reportedly exist in vivo not only as homotrimers, but also as BLyS/APRIL heterotrimers. Methods A proprietary N-terminal trimerization domain was used to produce recombinant BLyS/APRIL heterotrimers. Heterotrimer biologic activity was compared with that of BLyS and APRIL in a 4-hour signaling assay by using transmembrane activator and CAML interactor (TACI)-transfected Jurkat cells and in a 4-day primary human B-cell proliferation assay. A bead-based immunoassay was developed to quantify native heterotrimers in human sera from healthy donors (n = 89) and patients with systemic lupus erythematosus (SLE; n = 89) or rheumatoid arthritis (RA; n = 30). Heterotrimer levels were compared with BLyS and APRIL homotrimer levels in a subset of these samples. Results The recombinant heterotrimers consisted mostly of one BLyS and two APRIL molecules. Heterotrimer signaling did not show any significant difference compared with APRIL in the TACI-Jurkat assay. Heterotrimers were less-potent inducers of B-cell proliferation than were homotrimeric BLyS or APRIL (EC50, nMol/L: BLyS, 0.02; APRIL, 0.17; heterotrimers, 4.06). The soluble receptor fusion proteins atacicept and B-cell maturation antigen (BCMA)-immunoglobulin (Ig) neutralized the activity of BLyS, APRIL, and heterotrimers in both cellular assays, whereas B-cell activating factor belonging to the TNF family receptor (BAFF-R)-Ig neutralized only the activity of BLyS. In human sera, significantly more patients with SLE had detectable BLyS (67% versus 18%; P < 0.0001), APRIL (38% versus 3%; P < 0.0002), and heterotrimer (27% versus 8%; P = 0.0013) levels compared with healthy donors. Significantly more patients with RA had detectable APRIL, but not BLyS or heterotrimer, levels compared with healthy donors (83% versus 3%; P < 0.0001). Heterotrimer levels weakly correlated with BLyS, but not APRIL, levels. Conclusions Recombinant BLyS/APRIL heterotrimers have biologic activity and are inhibited by atacicept and BCMA-Ig, but not by BAFF-R-Ig. A novel immunoassay demonstrated that native BLyS/APRIL heterotrimers, as well as BLyS and APRIL homotrimers, are elevated in patients with autoimmune diseases.
Collapse
Affiliation(s)
- Stacey R Dillon
- Preclinical Research and Development, ZymoGenetics, Inc, 1201 Eastlake Ave East, Seattle, WA 98102, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Sun B, Wang H, Wang X, Huang H, Ding W, Jing R, Shi G, Zhu L. A proliferation-inducing ligand: a new biomarker for non-small cell lung cancer. Exp Lung Res 2010; 35:486-500. [PMID: 19842833 DOI: 10.1080/01902140902759274] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
To identify a proliferation-inducing ligand (APRIL) expression profile in tumor tissue and sputum of lung cancer, and evaluate the possibility of an assistant diagnosis of lung cancer by real-time fluorescence quantitative polymerase chain reaction (RTQ-PCR) in sputum as well, the authors analyzed the expression of APRIL mRNA in 75 tissue samples and 71 corresponding sputum samples of lung cancer by RTQ-PCR and analyzed their correlation. APRIL protein expression was also observed in tumor tissues by Western blot and immunohistochemistry. The expression analysis revealed APRIL expression was elevated in non-small cell lung cancer (NSCLC) and the expression of APRIL protein was located in the membrane and cytoplasm of tumor cells by immunohistochemiscal staining. Compared to benign pulmonary disease and healthy volunteers, the expression of APRIL mRNA in sputum of lung cancer was elevated (both P <. 001). When cut-off values for positivity were set at the mean + 2SD of mRNA expression in healthy volunteers, the positive rate for APRIL mRNA expression was 81.7% (58/71) in sputum samples of lung cancer, 3.2% (2/62) in benign pulmonary disease, and 1.5% (1/65) in healthy volunteers. The correlation was evident between the expression level of APRIL mRNA of tissue samples and that of sputum samples (P <. 001, r =. 702). These results support the possibility that the APRIL gene may play a key role in lung cancer, especially in NSCLC. The elevated expression level of APRIL mRNA in sputum of NSCLC suggested that APRIL mRNA may serve as an effective and convenient diagnostic biomarker for NSCLC.
Collapse
Affiliation(s)
- Baolan Sun
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Jiangsu Province, China
| | | | | | | | | | | | | | | |
Collapse
|
60
|
APRIL knockdown suppresses migration and invasion of human colon carcinoma cells. Clin Biochem 2009; 42:1694-8. [DOI: 10.1016/j.clinbiochem.2009.06.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2009] [Revised: 06/17/2009] [Accepted: 06/18/2009] [Indexed: 01/05/2023]
|
61
|
Gabay C, Krenn V, Bosshard C, Seemayer CA, Chizzolini C, Huard B. Synovial tissues concentrate secreted APRIL. Arthritis Res Ther 2009; 11:R144. [PMID: 19788740 PMCID: PMC2787289 DOI: 10.1186/ar2817] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Revised: 09/09/2009] [Accepted: 09/29/2009] [Indexed: 01/10/2023] Open
Abstract
INTRODUCTION A proliferation-inducing ligand (APRIL) from the TNF family, owing to its role in the generation and survival of plasma cells (PCs), is currently targeted for rheumatoid arthritis (RA) treatment. However, little is known about APRIL expression in RA lesions, hampering our understanding of the way APRIL may modulate this autoimmune disease. METHODS We performed immunological staining of human normal, non-RA and RA synovial tissues with a pair of antibodies specifically recognizing APRIL-producing cells and secreted APRIL. RESULTS We detected significant amounts of secreted APRIL in normal synovium mostly concentrated around blood vessels and at the lining layer, but no cells producing APRIL. Meanwhile, we observed that blood neutrophils constitutively secrete APRIL, indicating that blood APRIL may diffuse into the synovium via its fenestrated vessels. Synovium from non-RA and RA patients retained similarly secreted APRIL, but in this case APRIL-producing cells, including neutrophils and macrophages, were present in the tissue. Notably, PCs--when present in RA synovium--accumulated in areas of APRIL retention, spreading from blood vessels towards the lining layer. CONCLUSIONS PCs accumulate in synovial zones rich in secreted APRIL, consistent with a pro-survival role of APRIL for PCs in RA. The concentration of APRIL by normal synovium indicates that this tissue may constitute a proper environment for PCs even before RA onset.
Collapse
Affiliation(s)
- Cem Gabay
- Division of Rheumatology, University Hospitals, 4 rue Gabrielle Perret-Gentil, Geneva, CH 1211, Switzerland.
| | | | | | | | | | | |
Collapse
|
62
|
Le Pottier L, Bendaoud B, Renaudineau Y, Youinou P, Pers JO, Daridon C. New ELISA for B cell-activating factor. Clin Chem 2009; 55:1843-51. [PMID: 19696115 DOI: 10.1373/clinchem.2009.129940] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND The B cell-activating factor of the TNF family (BAFF) is upregulated in autoimmune diseases, but a number of conflicting results have cast doubts on the reliability of the ELISA protocols currently used for its quantification. This situation led us to develop a new ELISA for the measurement of BAFF. METHODS BAFF was purified for use alongside nonglycosylated recombinant BAFF. Two monoclonal antibodies (mAbs) and two polyclonal antibodies (pAbs) to BAFF were used. RESULTS The optimization process showed that the pAb format was preferable to the mAb format as capture antibody, because the pAbs recognized the glycosylated as well as the nonglycosylated forms of BAFF. The most efficient pair of Abs involved using the unconjugated form of a goat pAb to capture BAFF and the same biotinylated goat pAb to detect bound BAFF. This ELISA was not influenced by the presence of rheumatoid factor. CONCLUSIONS This new ELISA helped provide insights into why serum concentrations of BAFF vary between studies for a given population of patients. It is a reliable tool for the management of the diseases in which BAFF is an indication of response to therapy.
Collapse
Affiliation(s)
- Laëtitia Le Pottier
- EA 2216 Immunologie et Pathologie and IFR 418 ScInBioS, Université de Brest, Brest, France
| | | | | | | | | | | |
Collapse
|
63
|
Park M, Jung S, Park Y, Lee S. Relationship of serum TWEAK level to cytokine level, disease activity, and response to anti‐TNF treatment in patients with rheumatoid arthritis. Scand J Rheumatol 2009; 37:173-8. [DOI: 10.1080/03009740801898608] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
64
|
Rickman DS, Pflueger D, Moss B, VanDoren VE, Chen CX, de la Taille A, Kuefer R, Tewari AK, Setlur SR, Demichelis F, Rubin MA. SLC45A3-ELK4 is a novel and frequent erythroblast transformation-specific fusion transcript in prostate cancer. Cancer Res 2009; 69:2734-8. [PMID: 19293179 DOI: 10.1158/0008-5472.can-08-4926] [Citation(s) in RCA: 166] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Chromosomal rearrangements account for all erythroblast transformation-specific (ETS) family member gene fusions that have been reported in prostate cancer and have clinical, diagnostic, and prognostic implications. Androgen-regulated genes account for the majority of the 5' genomic regulatory promoter elements fused with ETS genes. TMPRSS2-ERG, TMPRSS2-ETV1, and SLC45A3-ERG rearrangements account for roughly 90% of ETS fusion prostate cancer. ELK4, another ETS family member, is androgen regulated, involved in promoting cell growth, and highly expressed in a subset of prostate cancer, yet the mechanism of ELK4 overexpression is unknown. In this study, we identified a novel ETS family fusion transcript, SLC45A3-ELK4, and found it to be expressed in both benign prostate tissue and prostate cancer. We found high levels of SLC45A3-ELK4 mRNA restricted to a subset of prostate cancer samples. SLC45A3-ELK4 transcript can be detected at high levels in urine samples from men at risk for prostate cancer. Characterization of the fusion mRNA revealed a major variant in which SLC45A3 exon 1 is fused to ELK4 exon 2. Based on quantitative PCR analyses of DNA, unlike other ETS fusions described in prostate cancer, the expression of SLC45A3-ELK4 mRNA is not exclusive to cases harboring a chromosomal rearrangement. Treatment of LNCaP cancer cells with a synthetic androgen (R1881) revealed that SLC45A3-ELK4, and not endogenous ELK4, mRNA expression is androgen regulated. Altogether, our findings show that SLC45A3-ELK4 mRNA expression is heterogeneous, highly induced in a subset of prostate cancers, androgen regulated, and most commonly occurs through a mechanism other than chromosomal rearrangement (e.g., trans-splicing).
Collapse
Affiliation(s)
- David S Rickman
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York 10021, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Moreaux J, Veyrune JL, De Vos J, Klein B. APRIL is overexpressed in cancer: link with tumor progression. BMC Cancer 2009; 9:83. [PMID: 19291294 PMCID: PMC2662875 DOI: 10.1186/1471-2407-9-83] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2008] [Accepted: 03/16/2009] [Indexed: 12/27/2022] Open
Abstract
Background BAFF and APRIL share two receptors – TACI and BCMA – and BAFF binds to a third receptor, BAFF-R. Increased expression of BAFF and APRIL is noted in hematological malignancies. BAFF and APRIL are essential for the survival of normal and malignant B lymphocytes, and altered expression of BAFF or APRIL or of their receptors (BCMA, TACI, or BAFF-R) have been reported in various B-cell malignancies including B-cell non-Hodgkin's lymphoma, chronic lymphocytic leukemia, Hodgkin's lymphoma, multiple myeloma, and Waldenstrom's macroglobulinemia. Methods We compared the expression of BAFF, APRIL, TACI and BAFF-R gene expression in 40 human tumor types – brain, epithelial, lymphoid, germ cells – to that of their normal tissue counterparts using publicly available gene expression data, including the Oncomine Cancer Microarray database. Results We found significant overexpression of TACI in multiple myeloma and thyroid carcinoma and an association between TACI expression and prognosis in lymphoma. Furthermore, BAFF and APRIL are overexpressed in many cancers and we show that APRIL expression is associated with tumor progression. We also found overexpression of at least one proteoglycan with heparan sulfate chains (HS), which are coreceptors for APRIL and TACI, in tumors where APRIL is either overexpressed or is a prognostic factor. APRIL could induce survival or proliferation directly through HS proteoglycans. Conclusion Taken together, these data suggest that APRIL is a potential prognostic factor for a large array of malignancies.
Collapse
Affiliation(s)
- Jérôme Moreaux
- CHU Montpellier, Institute of Research in Biotherapy, Montpellier, France.
| | | | | | | |
Collapse
|
66
|
Kimberley FC, van Bostelen L, Cameron K, Hardenberg G, Marquart JA, Hahne M, Medema JP. The proteoglycan (heparan sulfate proteoglycan) binding domain of APRIL serves as a platform for ligand multimerization and cross-linking. FASEB J 2009; 23:1584-95. [PMID: 19141538 DOI: 10.1096/fj.08-124669] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
A proliferation-inducing ligand (APRIL) (also known as TALL-2 and TRDL-1) is a member of the tumor necrosis factor (TNF) superfamily that has tumorigenic properties but is also important for the induction of humoral immune responses. APRIL binds two TNF receptors: transmembrane activator and calcium modulator and cyclophilin ligand interactor (TACI) and B-cell maturation antigen (BCMA) as well as heparan sulfate proteoglycans (HSPGs). The aim of this study was to clarify the role of the HSPG interaction in canonical APRIL signaling, because it has been proposed to act as a docking site and also to play a role in direct signaling. In this study, we generated point mutants of soluble APRIL that lack either the capacity to bind HSPGs or TACI and BCMA and then tested the function of these mutants in mouse B-cell assays. In contrast to previous reports, we found that APRIL alone is sufficient to costimulate B-cell proliferation and drive IgA production and does not require artificial antibody cross-linking. We found no evidence that APRIL requires signaling through HSPGs but, notably, were able to show that binding of APRIL to HSPGs is crucial for mediating natural APRIL cross-linking to allow for optimal activation of murine B cells.
Collapse
Affiliation(s)
- Fiona C Kimberley
- Laboratory of Experimental Oncology and Radiobiology, Center for Experimental Molecular Medicine, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
67
|
Targeting of BAFF and APRIL for Autoimmunity and Oncology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 647:52-63. [DOI: 10.1007/978-0-387-89520-8_4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
68
|
Kimberley FC, Medema JP, Hahne M. APRIL in B-cell malignancies and autoimmunity. Results Probl Cell Differ 2009; 49:161-182. [PMID: 19142624 DOI: 10.1007/400_2008_19] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
A Proliferation Inducing Ligand (APRIL) was first identified as a cytokine expressed predominantly by tumour tissues and was not found in most normal tissues. The activity of this new cytokine, in terms of its ability to stimulate tumour cell proliferation in vivo, determined the catchy acronym of yet another TNF family cytokine: APRIL. Reports showing an association between APRIL and cancer have since been prolific, in particular, those showing a link with B cell malignancies. Evidence is accumulating that APRIL is also a player in several autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis, and Sjoegren's syndrome. However, we now know that APRIL also plays an important role in the immune system and in lymphocyte biology. In this chapter we outline the physiological role of APRIL in immunity and describe what is known regarding the role of APRIL in B cell malignancies and autoimmune disease.
Collapse
Affiliation(s)
- Fiona C Kimberley
- Lab of Exp. Oncology and Radiobiology, Center for Experimental Molecular Medicine, Academic Medical Center, University of Amsterdam, Room G1-114, 1105 AZ, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
69
|
Kimberley FC, Hahne M, Medema JP. "APRIL hath put a spring of youth in everything": Relevance of APRIL for survival. J Cell Physiol 2008; 218:1-8. [PMID: 18726990 DOI: 10.1002/jcp.21561] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A proliferation inducing ligand (APRIL or TALL-2 and TRDL-1) was first discovered as a cytokine over-expressed in many transformed cells and with the capacity to stimulate proliferation. APRIL was shown to bind two different receptors of the TNF receptor superfamily: B cell maturation antigen (BCMA) and transmembrane activator and calcium modulator and cyclophilin ligand interactor (TACI), as well as heparan sulphate proteoglycans (HSPGs). APRIL has since been shown to play a physiological role in B cell biology, in particular the survival of plasma B cells in a specialized APRIL-rich niche. However, aberrant expression of APRIL and the subsequent activation of pro-survival pathways, is potentially the driving force for the survival of several B cell malignancies. APRIL has therefore become an important therapeutic target, but many questions regarding its mechanism of action still remain. It is for instance unclear what the exact physiological implications of the APRIL-HSPG interaction could be. Neither do we know the precise signals elicited by APRIL in normal or in malignant cells, and whether blocking these effects could provide real therapeutic gain in cancer patients. In this review we discuss the specific relevance of APRIL for cell survival, in terms of both its physiological role and its role in tumor biology, and highlight some of the key questions that will undoubtedly form the basis of future research in this field.
Collapse
Affiliation(s)
- Fiona C Kimberley
- LEXOR (Lab of Experimental Oncology and Radiobiology), Center for Experimental Molecular Medicine, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | | | | |
Collapse
|
70
|
The BLyS family: toward a molecular understanding of B cell homeostasis. Cell Biochem Biophys 2008; 53:1-16. [PMID: 19034695 DOI: 10.1007/s12013-008-9036-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2008] [Indexed: 12/27/2022]
Abstract
The B Lymphocyte Stimulator (BLyS) family of ligands and receptors regulates humoral immunity by controlling B lymphocyte survival and differentiation. Herein, we review the ligands and receptors of this family, their biological functions, and the biochemical processes through which they operate. Pre-immune B lymphocytes rely on BLyS signaling for their survival, whereas antigen experienced B lymphocytes generally interact more avidly with a homologous cytokine, A Proliferation Inducing Ligand (APRIL). The molecular basis for signaling via the three BLyS family receptors reveals complex interplay with other B lymphocyte signaling systems, affording the integration of selective and homeostatic processes. As our understanding of this system advances, molecular targets for manipulating humoral immunity in both health and disease should be revealed.
Collapse
|
71
|
Calvanese V, Mallya M, Campbell RD, Aguado B. Regulation of expression of two LY-6 family genes by intron retention and transcription induced chimerism. BMC Mol Biol 2008; 9:81. [PMID: 18817541 PMCID: PMC2562388 DOI: 10.1186/1471-2199-9-81] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Accepted: 09/25/2008] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Regulation of the expression of particular genes can rely on mechanisms that are different from classical transcriptional and translational control. The LY6G5B and LY6G6D genes encode LY-6 domain proteins, whose expression seems to be regulated in an original fashion, consisting of an intron retention event which generates, through an early premature stop codon, a non-coding transcript, preventing expression in most cell lines and tissues. RESULTS The MHC LY-6 non-coding transcripts have shown to be stable and very abundant in the cell, and not subject to Nonsense Mediated Decay (NMD). This retention event appears not to be solely dependent on intron features, because in the case of LY6G5B, when the intron is inserted in the artificial context of a luciferase expression plasmid, it is fully spliced but strongly stabilises the resulting luciferase transcript. In addition, by quantitative PCR we found that the retained and spliced forms are differentially expressed in tissues indicating an active regulation of the non-coding transcript. EST database analysis revealed that these genes have an alternative expression pathway with the formation of Transcription Induced Chimeras (TIC). This data was confirmed by RT-PCR, revealing the presence of different transcripts that would encode the chimeric proteins CSNKbeta-LY6G5B and G6F-LY6G6D, in which the LY-6 domain would join to a kinase domain and an Ig-like domain, respectively. CONCLUSION In conclusion, the LY6G5B and LY6G6D intron-retained transcripts are not subjected to NMD and are more abundant than the properly spliced forms. In addition, these genes form chimeric transcripts with their neighbouring same orientation 5' genes. Of interest is the fact that the 5' genes (CSNKbeta or G6F) undergo differential splicing only in the context of the chimera (CSNKbeta-LY6G5B or G6F-LY6G6C) and not on their own.
Collapse
Affiliation(s)
- Vincenzo Calvanese
- Centro de Biología Molecular Severo Ochoa (CBMSO), CSIC, Madrid, 28804, Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid 28029, Spain
| | - Meera Mallya
- MRC Rosalind Franklin Centre for Genomics Research, Cambridge, CB10 1SB, UK
- Department of Medicine, University of Cambridge, Wolfson College, CB3 9BB, UK
| | - R Duncan Campbell
- MRC Rosalind Franklin Centre for Genomics Research, Cambridge, CB10 1SB, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3QX, UK
| | - Begoña Aguado
- Centro de Biología Molecular Severo Ochoa (CBMSO), CSIC, Madrid, 28804, Spain
- MRC Rosalind Franklin Centre for Genomics Research, Cambridge, CB10 1SB, UK
| |
Collapse
|
72
|
Shui Y, Guan ZB, Zhang SQ. Molecular characterization of cytokine TWEAK and its receptor Fn14 in pig (Sus scrofa). Vet Immunol Immunopathol 2008; 126:396-402. [PMID: 18817982 DOI: 10.1016/j.vetimm.2008.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2008] [Revised: 08/04/2008] [Accepted: 08/18/2008] [Indexed: 11/24/2022]
Abstract
Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) is a member of the tumor necrosis factor superfamily (TNFSF). The interaction of TWEAK with its receptor fibroblast growth factor-inducible 14 (Fn14) regulates multiple cellular responses, including stimulation of proliferation, migration, apoptosis, angiogenesis, and induction of proinflammatory cytokines. This paper reports for the first time the molecular cloning of porcine TWEAK and Fn14 by EST and RACE strategies. The full-length cDNA of porcine TWEAK is 1327bp, including an open reading frame (ORF) of 747bp. Its genomic DNA consists of seven exons and six introns and is approximately 10kb in size by computer-assisted analysis. Sequence similarity at the amino acid level between porcine TWEAK and human or mouse was 95 and 92%, respectively. The full-length cDNA of porcine Fn14 contains 691bp, of which 390bp are the ORF. Sequence similarity at the amino acid level between porcine Fn14 and human, or mouse, or frog was 95, 93 and 64%, respectively. Real-time quantitative PCR (Q-PCR) analysis revealed that both TWEAK and Fn14 are constitutively expressed in various tissues in pig. Our results suggest that the TWEAK-Fn14 pathway is evolutionarily highly conserved. It will be helpful for investigation on the biological role of the TWEAK/Fn14 system in this important animal model. Furthermore, it provides insight into the molecular evolution of the emerging TWEAK and Fn14 families.
Collapse
Affiliation(s)
- Yan Shui
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, Life Sciences College, Nanjing Normal University, Nanjing 210046, PR China
| | | | | |
Collapse
|
73
|
|
74
|
Kunarso G, Wong KY, Stanton LW, Lipovich L. Detailed characterization of the mouse embryonic stem cell transcriptome reveals novel genes and intergenic splicing associated with pluripotency. BMC Genomics 2008; 9:155. [PMID: 18400104 PMCID: PMC2375908 DOI: 10.1186/1471-2164-9-155] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2007] [Accepted: 04/09/2008] [Indexed: 12/31/2022] Open
Abstract
Background Transcriptional control of embryonic stem (ES) cell pluripotency has been a subject of intense study. Transcriptional regulators including Oct4 (Oct3/4 index), Sox2 and Nanog are fundamental for maintaining the undifferentiated state. However, the ES cell transcriptome is not limited to their targets, and exhibits considerable complexity when assayed with microarray, MPSS, cDNA/EST sequencing, and SAGE technologies. To identify novel genes associated with pluripotency, we globally searched for ES transcripts not corresponding to known genes, validated their sequences, determined their expression profiles, and employed RNAi to test their function. Results Gene Identification Signature (GIS) analysis, a SAGE derivative distinguished by paired 5' and 3' transcript end tags, identified 153 candidate novel transcriptional units (TUs) distinct from known genes in a mouse E14 ES mRNA library. We focused on 16 TUs free of artefacts and mapping discrepancies, five of which were validated by RTPCR product sequencing. Two of the TUs were revealed by annotation to represent novel protein-coding genes: a PRY-domain cluster member and a KRAB-domain zinc finger. The other three TUs represented intergenic splicing events involving adjacent, functionally unrelated protein-coding genes transcribed in the same orientation, with one event potentially encoding a fusion protein containing domains from both component genes (Clk2 and Scamp3). Expression profiling using embryonic samples and adult tissue panels confirmed that three of the TUs were unique to or most highly expressed in ES cells. Expression levels of all five TUs dropped dramatically during three distinct chemically induced differentiation treatments of ES cells in culture. However, siRNA knockdowns of the TUs did not alter mRNA levels of pluripotency or differentiation markers, and did not affect cell morphology. Conclusion Transcriptome libraries retain considerable potential for novel gene discovery despite massive recent cDNA and EST sequencing efforts; cDNA and EST evidence for these ES cell TUs had been limited or absent. RTPCR and full-length sequencing remain essential in resolving the bottleneck between numerous candidate novel transcripts inferred from high-throughput sequencing and the small fraction that can be validated. RNAi results indicate that, despite their strong association with pluripotency, these five transcriptomic novelties may not be required for maintaining it.
Collapse
Affiliation(s)
- Galih Kunarso
- Computational & Mathematical Biology, Genome Institute of Singapore, 60 Biopolis Street #02-01, Singapore 138672, Singapore.
| | | | | | | |
Collapse
|
75
|
Smirnova AS, Andrade-Oliveira V, Gerbase-DeLima M. Identification of new splice variants of the genes BAFF and BCMA. Mol Immunol 2008; 45:1179-83. [PMID: 17825416 DOI: 10.1016/j.molimm.2007.07.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2007] [Accepted: 07/27/2007] [Indexed: 01/01/2023]
Abstract
The TNF superfamily ligands BAFF and APRIL and receptors BCMA, TACI and BAFF-R play an important role in the regulation of B cell immunity. A number of functionally important splice isoforms have already been characterized for these molecules, stimulating the search for new transcript variants (TVs). Here we report two new BAFF TVs and three BCMA TVs, all potentially codifying new proteins. BAFF TVs were expressed in peripheral blood mononuclear cells (PBMC) of nearly all the individuals studied, decreasing in level when PBMC were activated by PMA and ionomycin. They were also detected in PBMC cytoplasmic RNA. Low levels of the BAFF TVs in all lymphocyte subpopulations analyzed suggest that their main source in PBMC are monocytes. BCMA TVs were observed only in some CD19+ cell samples. Functional studies concerning interaction between isoforms of BAFF, APRIL and their receptors are needed for elucidation of their significance in the immune response.
Collapse
Affiliation(s)
- Anna S Smirnova
- Immunogenetics Division, Department of Pediatrics, Universidade Federal de Sao Paulo, R. Loefgreen 1235, 04040-031 São Paulo, SP, Brazil.
| | | | | |
Collapse
|
76
|
Daridon C, Youinou P, Pers JO. BAFF, APRIL, TWE-PRIL: Who's who? Autoimmun Rev 2008; 7:267-71. [DOI: 10.1016/j.autrev.2007.05.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2007] [Accepted: 05/14/2007] [Indexed: 10/23/2022]
|
77
|
Wang K, Ubriaco G, Sutherland LC. RBM6-RBM5 transcription-induced chimeras are differentially expressed in tumours. BMC Genomics 2007; 8:348. [PMID: 17908320 PMCID: PMC2174484 DOI: 10.1186/1471-2164-8-348] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2007] [Accepted: 10/01/2007] [Indexed: 11/29/2022] Open
Abstract
Background Transcription-induced chimerism, a mechanism involving the transcription and intergenic splicing of two consecutive genes, has recently been estimated to account for ~5% of the human transcriptome. Despite this prevalence, the regulation and function of these fused transcripts remains largely uncharacterised. Results We identified three novel transcription-induced chimeras resulting from the intergenic splicing of a single RNA transcript incorporating the two neighbouring 3p21.3 tumour suppressor locus genes, RBM6 and RBM5, which encode the RNA Binding Motif protein 6 and RNA Binding Motif protein 5, respectively. Each of the three novel chimeric transcripts lacked exons 3, 6, 20 and 21 of RBM6 and exon 1 of RBM5. Differences between the transcripts were associated with the presence or absence of exon 4, exon 5 and a 17 nucleotide (nt) sequence from intron 10 of RBM6. All three chimeric transcripts incorporated the canonical splice sites from both genes (excluding the 17 nt intron 10 insertion). Differential expression was observed in tumour tissue compared to non-tumour tissue, and amongst tumour types. In breast tumour tissue, chimeric expression was associated with elevated levels of RBM6 and RBM5 mRNA, and increased tumour size. No protein expression was detected by in vitro transcription/translation. Conclusion These results suggest that RBM6 mRNA experiences altered co-transcriptional gene regulation in certain cancers. The results also suggest that RBM6-RBM5 transcription-induced chimerism might be a process that is linked to the tumour-associated increased transcriptional activity of the RBM6 gene. It appears that none of the transcription-induced chimeras generates a protein product; however, the novel alternative splicing, which affects putative functional domains within exons 3, 6 and 11 of RBM6, does suggest that the generation of these chimeric transcripts has functional relevance. Finally, the association of chimeric expression with breast tumour size suggests that RBM6-RBM5 chimeric expression may be a potential tumour differentiation marker.
Collapse
Affiliation(s)
- Ke Wang
- Tumour Biology Group, Regional Cancer Program of the Sudbury Regional Hospital, Sudbury, Ontario, Canada
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, Jilin, China
| | - Gino Ubriaco
- Northern Ontario School of Medicine, Sudbury, Ontario, Canada
| | - Leslie C Sutherland
- Tumour Biology Group, Regional Cancer Program of the Sudbury Regional Hospital, Sudbury, Ontario, Canada
- Northern Ontario School of Medicine, Sudbury, Ontario, Canada
- Biomolecular Sciences Program, Laurentian University, Sudbury, Ontario, Canada
| |
Collapse
|
78
|
TWEAK and the Central Nervous System. Mol Neurobiol 2007; 35:255-65. [DOI: 10.1007/s12035-007-0024-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2006] [Revised: 11/30/1999] [Accepted: 11/10/2006] [Indexed: 12/22/2022]
|
79
|
Hosokawa Y, Hosokawa I, Ozaki K, Nakae H, Matsuo T. Proinflammatory effects of tumour necrosis factor-like weak inducer of apoptosis (TWEAK) on human gingival fibroblasts. Clin Exp Immunol 2007; 146:540-9. [PMID: 17100776 PMCID: PMC1810398 DOI: 10.1111/j.1365-2249.2006.03233.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Tumour necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK), a member of the TNF family, is a multi-functional cytokine that regulates cellular proliferation, angiogenesis, inflammation and apoptosis. In this study, we investigated TWEAK expression in periodontally diseased tissues and the effect of TWEAK on human gingival fibroblasts (HGF). Reverse transcription-polymerase chain reaction (RT-PCR) analysis and immunohistochemistry revealed that TWEAK and the TWEAK receptor, fibroblast growth factor-inducible 14 (Fn14), mRNA and protein were expressed in periodontally diseased tissues. HGF expressed Fn14 and produced interleukin (IL)-8 and vascular endothelial growth factor (VEGF) production upon TWEAK stimulation in a dose-dependent manner. The IL-8 and VEGF production induced by TWEAK was augmented synergistically by simultaneous stimulation with transforming growth factor (TGF)-beta1 or IL-1beta. IL-1beta and TGF-beta1 enhanced Fn14 expression in a dose-dependent manner. Moreover, TWEAK induced intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) expression on HGF in a dose-dependent manner. The ICAM-1 expression induced by TWEAK was augmented by TGF-beta1. On the other hand, the TWEAK-induced VCAM-1 expression was inhibited by TGF-beta1. Phosphatidylinositol 3-kinase (PI3K) and nuclear factor-kappaB (NF-kappaB) inhibitor inhibit both ICAM-1 and VCAM-1 expression induced by TWEAK. However, mitogen-activated protein kinase (MEK) and c-Jun NH2-terminal kinase (JNK) inhibitor enhanced only VCAM-1 expression on HGF. These results suggest that TWEAK may be involved in the pathophysiology of periodontal disease. Moreover, in combination with IL-1beta or TGF-beta1, TWEAK may be related to the exacerbation of periodontal disease to induce proinflammatory cytokines and adherent molecules by HGF.
Collapse
Affiliation(s)
- Y Hosokawa
- Department of Conservative Dentistry and Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Tokushima, Japan.
| | | | | | | | | |
Collapse
|
80
|
Kwaśnicka-Crawford DA, Carson AR, Scherer SW. IQCJ-SCHIP1, a novel fusion transcript encoding a calmodulin-binding IQ motif protein. Biochem Biophys Res Commun 2006; 350:890-9. [PMID: 17045569 DOI: 10.1016/j.bbrc.2006.09.136] [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] [Received: 08/30/2006] [Accepted: 09/21/2006] [Indexed: 10/24/2022]
Abstract
The existence of transcripts that span two adjacent, independent genes is considered rare in the human genome. This study characterizes a novel human fusion gene named IQCJ-SCHIP1. IQCJ-SCHIP1 is the longest isoform of a complex transcriptional unit that bridges two separate genes that encode distinct proteins, IQCJ, a novel IQ motif containing protein and SCHIP1, a schwannomin interacting protein that has been previously shown to interact with the Neurofibromatosis type 2 (NF2) protein. IQCJ-SCHIP1 is located on the chromosome 3q25 and comprises a 1692-bp transcript encompassing 11 exons spanning 828kb of the genomic DNA. We show that IQCJ-SCHIP1 mRNA is highly expressed in the brain. Protein encoded by the IQCJ-SCHIP1 gene was localized to cytoplasm and actin-rich regions and in differentiated PC12 cells was also seen in neurite extensions.
Collapse
|
81
|
Chiu A, Xu W, He B, Dillon SR, Gross JA, Sievers E, Qiao X, Santini P, Hyjek E, Lee JW, Cesarman E, Chadburn A, Knowles DM, Cerutti A. Hodgkin lymphoma cells express TACI and BCMA receptors and generate survival and proliferation signals in response to BAFF and APRIL. Blood 2006; 109:729-39. [PMID: 16960154 PMCID: PMC1785096 DOI: 10.1182/blood-2006-04-015958] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Hodgkin lymphoma (HL) originates from the clonal expansion of malignant Hodgkin and Reed-Sternberg (HRS) cells. These B-cell-derived elements constitute less than 10% of the tumoral mass. The remaining tissue is comprised of an inflammatory infiltrate that includes myeloid cells. Myeloid cells activate B cells by producing BAFF and APRIL, which engage TACI, BCMA, and BAFF-R receptors on the B cells. Here, we studied the role of BAFF and APRIL in HL. Inflammatory and HRS cells from HL tumors expressed BAFF and APRIL. Unlike their putative germinal center B-cell precursors, HRS cells lacked BAFF-R, but expressed TACI and BCMA, a phenotype similar to that of plasmacytoid B cells. BAFF and APRIL enhanced HRS cell survival and proliferation by delivering nonredundant signals via TACI and BCMA receptors through both autocrine and paracrine pathways. These signals caused NF-kappaB activation; Bcl-2, Bcl-xL, and c-Myc up-regulation; and Bax down-regulation, and were amplified by APRIL-binding proteoglycans on HRS cells. Interruption of BAFF and APRIL signaling by TACI-Ig decoy receptor, which binds to and neutralizes BAFF and APRIL, or by small-interfering RNAs targeting BAFF, APRIL, TACI, and BCMA inhibited HRS cell accumulation in vitro and might attenuate HL expansion in vivo.
Collapse
Affiliation(s)
- April Chiu
- Department of Pathology and Laboratory Medicine of Weill Medical College of Cornell University, New York, NY
| | - Weifeng Xu
- Department of Pathology and Laboratory Medicine of Weill Medical College of Cornell University, New York, NY
| | - Bing He
- Department of Pathology and Laboratory Medicine of Weill Medical College of Cornell University, New York, NY
| | | | | | | | - Xugang Qiao
- Department of Pathology and Laboratory Medicine of Weill Medical College of Cornell University, New York, NY
| | - Paul Santini
- Department of Pathology and Laboratory Medicine of Weill Medical College of Cornell University, New York, NY
| | - Elizabeth Hyjek
- Department of Pathology and Laboratory Medicine of Weill Medical College of Cornell University, New York, NY
| | - Joong-won Lee
- Department of Pathology and Laboratory Medicine of Weill Medical College of Cornell University, New York, NY
| | - Ethel Cesarman
- Department of Pathology and Laboratory Medicine of Weill Medical College of Cornell University, New York, NY
- Graduate Program of Immunology and Microbial Pathogenesis, Weill Graduate School of Medical Sciences of Cornell University, New York, NY
| | - Amy Chadburn
- Department of Pathology and Laboratory Medicine of Weill Medical College of Cornell University, New York, NY
| | - Daniel M. Knowles
- Department of Pathology and Laboratory Medicine of Weill Medical College of Cornell University, New York, NY
| | - Andrea Cerutti
- Department of Pathology and Laboratory Medicine of Weill Medical College of Cornell University, New York, NY
- Graduate Program of Immunology and Microbial Pathogenesis, Weill Graduate School of Medical Sciences of Cornell University, New York, NY
- Correspondence: Andrea Cerutti,
Weill Medical College of Cornell University, Department of Pathology and Laboratory Medicine, 1300 York Ave, Rm C-410, New York, NY 10021; e-mail:
| |
Collapse
|
82
|
Abstract
BAFF is a key factor controlling B cell survival and maturation and its over-expression promotes B cell-mediated autoimmune disorders and participates in the progression of B cell lymphomas. Yet, BAFF and a related ligand APRIL are expressed by T lymphocytes and modulate their functions. BAFF and APRIL promote T cell activation and survival. BAFF over-expression in transgenic (Tg) mice enhances T helper 1 (Thl)-driven delayed-type hypersensitivity (DTH), but inhibits T helper 2 (Th2) cell-mediated allergic airway inflammation in mice. Some of these effects are also dependent on BAFF-induced modification of the B cell compartment. Therefore, direct BAFF/APRIL signalling in T cells and/or T cell modulation in response to a BAFF-modified B cell compartment may play an important role in inflammation and immunomodulation.
Collapse
Affiliation(s)
- Fabienne Mackay
- Department of Immunology and Inflammation, The Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.
| | | |
Collapse
|
83
|
Bossen C, Schneider P. BAFF, APRIL and their receptors: structure, function and signaling. Semin Immunol 2006; 18:263-75. [PMID: 16914324 DOI: 10.1016/j.smim.2006.04.006] [Citation(s) in RCA: 382] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2006] [Accepted: 04/03/2006] [Indexed: 12/17/2022]
Abstract
BAFF, APRIL and their receptors play important immunological roles, especially in the B cell arm of the immune system. A number of splice isoforms have been described for both ligands and receptors in this subfamily, some of which are conserved between mouse and human, while others are species-specific. Structural and mutational analyses have revealed key determinants of receptor-ligand specificity. BAFF-R has a strong selectivity for BAFF; BCMA has a higher affinity for APRIL than for BAFF, while TACI binds both ligands equally well. The molecular signaling events downstream of BAFF-R, BCMA and TACI are still incompletely characterized. Survival appears to be mediated by upregulation of Bcl-2 family members through NF-kappaB activation, degradation of the pro-apototic Bim protein, and control of subcellular localization of PCKdelta. Very little is known about other signaling events associated with receptor engagement by BAFF and APRIL that lead for example to B cell activation or to CD40L-independent Ig switch.
Collapse
Affiliation(s)
- Claudia Bossen
- Biochemistry Department, University of Lausanne, Boveresses 155, CH-1066 Epalinges, Switzerland
| | | |
Collapse
|
84
|
Hernández-Sánchez C, Bártulos O, Valenciano AI, Mansilla A, de Pablo F. The regulated expression of chimeric tyrosine hydroxylase-insulin transcripts during early development. Nucleic Acids Res 2006; 34:3455-64. [PMID: 16840532 PMCID: PMC1524912 DOI: 10.1093/nar/gkl436] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Biological complexity does not appear to be simply correlated with gene number but rather other mechanisms contribute to the morphological and functional diversity across phyla. Such mechanisms regulate different transcriptional, translational and post-translational processes and include the recently identified transcription induced chimerism (TIC). We have found two novel chimeric transcripts in the chick and quail that result from the fusion of tyrosine hydroxylase (TH) and insulin into a single mature transcript. The th and insulin genes are located in tandem and they are generally transcribed independently. However, it appears that two chimeric transcripts containing exons from both the genes can also be produced in a regulated manner. The TH–INS1 and TH–INS2 chimeras differ in their insulin gene content, and they encode two novel isoforms of the TH protein with markedly reduced functionality when compared with the canonical TH. In addition, the TH–INS1 chimeric mRNA generates a small amount of insulin. We propose that TIC is an additional mechanism that can be employed to further regulate TH and insulin expression according to the specific needs of developing vertebrates.
Collapse
Affiliation(s)
- Catalina Hernández-Sánchez
- Group of Growth Factors in Vertebrate Development, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Ramiro de Maeztu 9, E-28040 Madrid, Spain.
| | | | | | | | | |
Collapse
|
85
|
Miller JP, Stadanlick JE, Cancro MP. Space, selection, and surveillance: setting boundaries with BLyS. THE JOURNAL OF IMMUNOLOGY 2006; 176:6405-10. [PMID: 16709796 DOI: 10.4049/jimmunol.176.11.6405] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The BLyS family of ligands and receptors governs B cell homeostasis by controlling survival, differentiation, and lifespan. This family consists of multiple receptors and ligands, allowing independent regulation of different B cell subsets by varying the combination and levels of receptors expressed. Multiple downstream signaling pathways are implicated in these activities, reflecting this receptor complexity as well as cross-talk with other B cell signaling systems. BLyS levels are associated with multiple forms of humoral autoimmunity and can modulate tolerogenic elimination at the transitional checkpoint. BLyS responsiveness thus balances peripheral selection against cell numbers, providing an elastic system that varies selective stringency based on homeostatic demands.
Collapse
Affiliation(s)
- Juli P Miller
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
86
|
Dillon SR, Gross JA, Ansell SM, Novak AJ. An APRIL to remember: novel TNF ligands as therapeutic targets. Nat Rev Drug Discov 2006; 5:235-46. [PMID: 16474316 DOI: 10.1038/nrd1982] [Citation(s) in RCA: 233] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Since their discovery in 1998, the two TNF family members APRIL and BLyS/BAFF have received increasing attention. In addition to regulating normal B-cell development and immune responses, these molecules might be crucial in a diverse set of diseases, including autoimmunity and cancer. Although more has been published about the general biology of BLyS/BAFF than that of APRIL, many recent articles have described novel APRIL biology. Here we focus on APRIL, exploring its normal and pathological functions, and comparing the therapeutic molecules currently under development that target BLyS/BAFF alone, or APRIL and BLyS/BAFF together.
Collapse
Affiliation(s)
- Stacey R Dillon
- Department of Autoimmunity and Inflammation, ZymoGenetics, Inc. 1201 Eastlake Avenue East, Seattle, Washington 98102, USA.
| | | | | | | |
Collapse
|
87
|
Winkles JA, Tran NL, Berens ME. TWEAK and Fn14: New molecular targets for cancer therapy? Cancer Lett 2006; 235:11-7. [PMID: 15885893 DOI: 10.1016/j.canlet.2005.03.048] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2005] [Accepted: 03/24/2005] [Indexed: 11/24/2022]
Abstract
Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) is a member of the tumor necrosis factor (TNF) superfamily of structurally related cytokines. Full-length, membrane-anchored TWEAK can be found on the surface of many cell types and a smaller, biologically active form, generated via proteolytic processing, has also been detected in the extracellular milieu. TWEAK acts via binding to a recently identified TNF receptor superfamily member named fibroblast growth factor-inducible 14 (Fn14). It has been demonstrated that TWEAK binding to the Fn14 receptor, or constitutive Fn14 overexpression, activates the nuclear factor-kappaB signaling pathway, which is known to play an important role in immune and inflammatory processes, oncogenesis, and cancer therapy resistance. In this article, we review recent studies indicating that TWEAK and Fn14 may be potential regulators of human tumorigenesis.
Collapse
Affiliation(s)
- Jeffrey A Winkles
- Department of Surgery, University of Maryland Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, 21201, USA.
| | | | | |
Collapse
|
88
|
Valentijn LJ, Koster J, Versteeg R. Read-through transcript from NM23-H1 into the neighboring NM23-H2 gene encodes a novel protein, NM23-LV. Genomics 2006; 87:483-9. [PMID: 16442775 DOI: 10.1016/j.ygeno.2005.11.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2005] [Revised: 11/08/2005] [Accepted: 11/15/2005] [Indexed: 10/25/2022]
Abstract
NM23-H1 and NM23-H2 are neighboring genes on chromosome 17q. They encode nucleoside diphosphate kinases that have additional roles in signal transduction, transcription, and apoptosis. NM23-H1 expression is a strong marker for prognosis and metastatic behavior in many tumor types. A new bioinformatic tool, TranscriptView, identified read-through transcripts that start in the NM23-H1 gene and continue in the neighboring NM23-H2 gene. Splicing results in a transcript containing exons 1 to 4 of NM23-H1 and exons 2 to 5 of NM23-H2. The resulting mRNA encodes a novel and long variant of the NM23 protein family, NM23-LV, which contains part of NM23-H1 and the complete NM23-H2 protein. The transcript was amplified and sequenced from two neuroblastoma cell lines, confirming the presence of the predicted NM23-LV mRNA in vivo. Tissue analysis showed that NM23-LV is ubiquitously expressed, with the exception of the kidney. Neuroblastoma tumors show high-level expression of NM23-H1 and-H2 as well as NM23-LV mRNA. In neuroblastoma cells, the NM23-LV protein has mainly a cytoplasmic localization, but some nuclear staining was observed as well.
Collapse
MESH Headings
- Alternative Splicing
- Amino Acid Sequence
- Biomarkers, Tumor
- Blotting, Western
- Cell Line, Tumor
- Cell Nucleus/metabolism
- Chromosomes, Human, Pair 17
- Computational Biology
- Cytoplasm/metabolism
- DNA, Neoplasm/genetics
- Exons
- Expressed Sequence Tags
- Fluorescent Antibody Technique
- Fluorescent Dyes
- Gene Expression Regulation, Neoplastic
- Genetic Variation
- Humans
- Introns
- Molecular Sequence Data
- NM23 Nucleoside Diphosphate Kinases
- Neuroblastoma/genetics
- Nucleic Acid Amplification Techniques
- Nucleoside-Diphosphate Kinase/genetics
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Rhodamines
- Sequence Analysis, DNA
- Sequence Homology, Amino Acid
- Tissue Distribution
- Transcription, Genetic
Collapse
Affiliation(s)
- Linda J Valentijn
- Department of Human Genetics, M1-134, Academic Medical Center, University of Amsterdam, P.O. Box 22700, 1100 DE Amsterdam, The Netherlands.
| | | | | |
Collapse
|
89
|
Bossen C, Ingold K, Tardivel A, Bodmer JL, Gaide O, Hertig S, Ambrose C, Tschopp J, Schneider P. Interactions of tumor necrosis factor (TNF) and TNF receptor family members in the mouse and human. J Biol Chem 2006; 281:13964-71. [PMID: 16547002 DOI: 10.1074/jbc.m601553200] [Citation(s) in RCA: 339] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Ligands of the tumor necrosis factor superfamily (TNFSF) (4-1BBL, APRIL, BAFF, CD27L, CD30L, CD40L, EDA1, EDA2, FasL, GITRL, LIGHT, lymphotoxin alpha, lymphotoxin alphabeta, OX40L, RANKL, TL1A, TNF, TWEAK, and TRAIL) bind members of the TNF receptor superfamily (TNFRSF). A comprehensive survey of ligand-receptor interactions was performed using a flow cytometry-based assay. All ligands engaged between one and five receptors, whereas most receptors only bound one to three ligands. The receptors DR6, RELT, TROY, NGFR, and mouse TNFRH3 did not interact with any of the known TNFSF ligands, suggesting that they either bind other types of ligands, function in a ligand-independent manner, or bind ligands that remain to be identified. The study revealed that ligand-receptor pairs are either cross-reactive between human and mouse (e.g. Tweak/Fn14, RANK/RANKL), strictly species-specific (GITR/GITRL), or partially species-specific (e.g. OX40/OX40L, CD40/CD40L). Interestingly, the receptor binding patterns of lymphotoxin alpha and alphabeta are redundant in the human but not in the mouse system. Ligand oligomerization allowed detection of weak interactions, such as that of human TNF with mouse TNFR2. In addition, mouse APRIL exists as two different splice variants differing by a single amino acid. Although human APRIL does not interact with BAFF-R, the shorter variant of mouse APRIL exhibits weak but detectable binding to mouse BAFF-R.
Collapse
Affiliation(s)
- Claudia Bossen
- Biochemistry Department, University of Lausanne, CH-1066 Epalinges, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Parra G, Reymond A, Dabbouseh N, Dermitzakis ET, Castelo R, Thomson TM, Antonarakis SE, Guigó R. Tandem chimerism as a means to increase protein complexity in the human genome. Genes Dev 2006; 16:37-44. [PMID: 16344564 PMCID: PMC1356127 DOI: 10.1101/gr.4145906] [Citation(s) in RCA: 166] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2005] [Accepted: 09/28/2005] [Indexed: 11/24/2022]
Abstract
The "one-gene, one-protein" rule, coined by Beadle and Tatum, has been fundamental to molecular biology. The rule implies that the genetic complexity of an organism depends essentially on its gene number. The discovery, however, that alternative gene splicing and transcription are widespread phenomena dramatically altered our understanding of the genetic complexity of higher eukaryotic organisms; in these, a limited number of genes may potentially encode a much larger number of proteins. Here we investigate yet another phenomenon that may contribute to generate additional protein diversity. Indeed, by relying on both computational and experimental analysis, we estimate that at least 4%-5% of the tandem gene pairs in the human genome can be eventually transcribed into a single RNA sequence encoding a putative chimeric protein. While the functional significance of most of these chimeric transcripts remains to be determined, we provide strong evidence that this phenomenon does not correspond to mere technical artifacts and that it is a common mechanism with the potential of generating hundreds of additional proteins in the human genome.
Collapse
Affiliation(s)
- Genís Parra
- Grup de Recerca en Informàtica Biomèdica, Institut Municipal d'Investigació Mèdica-Universitat Pompeu Fabra, and Programa de Bioinformàtica i Genòmica, Centre de Regulació Genòmica, E08003 Barcelona, Catalonia, Spain
| | | | | | | | | | | | | | | |
Collapse
|
91
|
Akiva P, Toporik A, Edelheit S, Peretz Y, Diber A, Shemesh R, Novik A, Sorek R. Transcription-mediated gene fusion in the human genome. Genome Res 2005; 16:30-6. [PMID: 16344562 PMCID: PMC1356126 DOI: 10.1101/gr.4137606] [Citation(s) in RCA: 214] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Transcription of a gene usually ends at a regulated termination point, preventing the RNA-polymerase from reading through the next gene. However, sporadic reports suggest that chimeric transcripts, formed by transcription of two consecutive genes into one RNA, can occur in human. The splicing and translation of such RNAs can lead to a new, fused protein, having domains from both original proteins. Here, we systematically identified over 200 cases of intergenic splicing in the human genome (involving 421 genes), and experimentally demonstrated that at least half of these fusions exist in human tissues. We showed that unique splicing patterns dominate the functional and regulatory nature of the resulting transcripts, and found intergenic distance bias in fused compared with nonfused genes. We demonstrate that the hundreds of fused genes we identified are only a subset of the actual number of fused genes in human. We describe a novel evolutionary mechanism where transcription-induced chimerism followed by retroposition results in a new, active fused gene. Finally, we provide evidence that transcription-induced chimerism can be a mechanism contributing to the evolution of protein complexes.
Collapse
|
92
|
Salzer U, Grimbacher B. TACItly changing tunes: farewell to a yin and yang of BAFF receptor and TACI in humoral immunity? Curr Opin Allergy Clin Immunol 2005; 5:496-503. [PMID: 16264328 DOI: 10.1097/01.all.0000191887.89773.cc] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW The complex system of the tumour necrosis factor ligands BAFF and APRIL and their three receptors BCMA, TACI and BAFF receptor and its role in B-cell development and function is the objective of extensive research. Whereas the importance of BAFF/BAFF receptor interactions for B-cell survival could be clearly demonstrated, TACI is believed to counteract BAFF activity as a negative regulator in the murine model. The primarily immunodeficient phenotype of human TACI deficiency, however, claims a distinct function of this receptor in human peripheral B-cell development, class switch recombination and terminal differentiation. RECENT FINDINGS Common variable immunodeficiency comprises a heterogeneous group of antibody deficiency syndromes characterized by impaired terminal B-cell differentiation. By means of molecular genetics common variable immunodeficiency is still ill-defined, but the description of the deficiency of the inducible costimulator in a small subgroup of common variable immunodeficiency patients set the starting point for the molecular dissection of this disease entity. The recent discovery of genetic defects in the tumour necrosis factor receptor superfamily members TACI and BAFF receptor in patients with common variable immunodeficiency denotes further advances in this field. SUMMARY In this review we will discuss recent progress made in the understanding of the BAFF/APRIL-TACI/BCMA/BAFF receptor system in relation to the recent discovery that mutations in human TACI cause a primary humoral immunodeficiency. This suggests a refined role for TACI in human B-cell biology.
Collapse
Affiliation(s)
- Ulrich Salzer
- Department of Clinical Immunology and Rheumatology, Medical Center, University of Freiburg, Freiburg, Germany
| | | |
Collapse
|
93
|
Thangarajh M, Masterman T, Rot U, Duvefelt K, Brynedal B, Karrenbauer VD, Hillert J. Increased levels of APRIL (a proliferation-inducing ligand) mRNA in multiple sclerosis. J Neuroimmunol 2005; 167:210-4. [PMID: 16087247 DOI: 10.1016/j.jneuroim.2005.06.024] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2005] [Accepted: 06/27/2005] [Indexed: 11/15/2022]
Abstract
B cells play an indispensable, yet indeterminate, role in the pathogenesis of multiple sclerosis (MS). We measured mRNA of APRIL-a promotor of B-cell survival-in peripheral blood and quantified protein levels in plasma and cerebrospinal fluid in MS patients and controls. APRIL mRNA levels in monocytes and T cells were significantly higher in MS patients than in controls. Levels of soluble APRIL in plasma were higher in patients with chronic progressive MS than in patients with relapsing-remitting MS, albeit not significantly. MS may thus be associated with increased transcription in peripheral blood of factors promoting B-cell survival, including APRIL.
Collapse
Affiliation(s)
- Mathula Thangarajh
- Division of Neurology, Department of Clinical Neuroscience, Karolinska Institutet, Karolinska University Hospital, SE-141 86 Stockholm, Sweden.
| | | | | | | | | | | | | |
Collapse
|
94
|
Abstract
The TNF family ligands BAFF (also called BLyS) and APRIL regulate lymphocyte survival and activation. BAFF binds to three receptors, BAFF-R, TACI and BCMA, whereas APRIL interacts with TACI, BCMA and proteoglycans. The contribution of BAFF and APRIL to B-cell and plasma-cell survival, CD154 (CD40L)-independent antibody isotype switching, germinal center maintenance, T-dependent and T-independent antibody responses, and T cell co-stimulation are relatively well understood. Constitutive BAFF produced by stromal cells determines the size of the peripheral B cell pool, whereas inducible BAFF produced by myeloid and other cells supports local survival of B lymphocytes and can be associated with development of autoimmunity when deregulated.
Collapse
Affiliation(s)
- Pascal Schneider
- Biochemistry Department, University of Lausanne, Boveresses 155, CH-1066 Epalinges, Switzerland.
| |
Collapse
|
95
|
Ch'en PFT, Xu XG, Liu XS, Liu Y, Song CJ, Screaton GR, Jin BQ, Xu XN. Characterisation of monoclonal antibodies to the TNF and TNF receptor families. Cell Immunol 2005; 236:78-85. [PMID: 16157320 DOI: 10.1016/j.cellimm.2005.08.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2005] [Accepted: 05/06/2005] [Indexed: 12/16/2022]
Abstract
Tumour necrosis factor (TNF) family ligands and their corresponding receptors play important roles in the immune system and are involved in immune regulation such as lymphoid development, cell proliferation, differentiation, activation and death. Antibodies against these ligands and receptors together with Fc-fusion proteins, have been particularly useful as immunological tools in addressing the underlying involvement of these proteins in these contexts and furthermore, have given us hope in using them as potential therapeutic agents. Over last few years, there have been many additions to these ever-growing TNF family ligands and their receptors. Here, we have generated and characterised a set of monoclonal antibodies, together with mAbs from the HLDA workshop, against DcR1, DcR2, DR4, DR5, TRAIL, APRIL, BAFF, BAFF-R, BCMA, and TACI, which may be useful in phenotypic and functional studies of the role of TNF and TNF receptor family in immune function and regulation in relation to health and disease.
Collapse
Affiliation(s)
- Paul F-T Ch'en
- Weatherall Institute of Molecular Medicine, MRC Human Immunology Unit, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | | | | | | | | | | | | | | |
Collapse
|
96
|
Ingold K, Zumsteg A, Tardivel A, Huard B, Steiner QG, Cachero TG, Qiang F, Gorelik L, Kalled SL, Acha-Orbea H, Rennert PD, Tschopp J, Schneider P. Identification of proteoglycans as the APRIL-specific binding partners. ACTA ACUST UNITED AC 2005; 201:1375-83. [PMID: 15851487 PMCID: PMC2213192 DOI: 10.1084/jem.20042309] [Citation(s) in RCA: 269] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
B cell activating factor of the tumor necrosis factor (TNF) family (BAFF) and a proliferation-inducing ligand (APRIL) are closely related ligands within the TNF superfamily that play important roles in B lymphocyte biology. Both ligands share two receptors—transmembrane activator and calcium signal–modulating cyclophilin ligand interactor (TACI) and B cell maturation antigen (BCMA)—that are predominantly expressed on B cells. In addition, BAFF specifically binds BAFF receptor, whereas the nature of a postulated APRIL-specific receptor remains elusive. We show that the TNF homology domain of APRIL binds BCMA and TACI, whereas a basic amino acid sequence (QKQKKQ) close to the NH2 terminus of the mature protein is required for binding to the APRIL-specific “receptor.” This interactor was identified as negatively charged sulfated glycosaminoglycan side chains of proteoglycans. Although T cell lines bound little APRIL, the ectopic expression of glycosaminoglycan-rich syndecans or glypicans conferred on these cells a high binding capacity that was completely dependent on APRIL's basic sequence. Moreover, syndecan-1–positive plasma cells and proteoglycan-rich nonhematopoietic cells displayed high specific, heparin-sensitive binding to APRIL. Inhibition of BAFF and APRIL, but not BAFF alone, prevented the survival and/or the migration of newly formed plasma cells to the bone marrow. In addition, costimulation of B cell proliferation by APRIL was only effective upon APRIL oligomerization. Therefore, we propose a model whereby APRIL binding to the extracellular matrix or to proteoglycan-positive cells induces APRIL oligomerization, which is the prerequisite for the triggering of TACI- and/or BCMA-mediated activation, migration, or survival signals.
Collapse
Affiliation(s)
- Karine Ingold
- Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Cancro MP. The BLyS family of ligands and receptors: an archetype for niche-specific homeostatic regulation. Immunol Rev 2005; 202:237-49. [PMID: 15546397 DOI: 10.1111/j.0105-2896.2004.00212.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Discovery and characterization of the tumor necrosis factor (TNF) family member B-lymphocyte stimulator (BLyS) has opened a novel chapter in the role of TNF family members in the homeostatic control of lymphocyte populations. BLyS and its sister cytokine APRIL (a proliferation-inducing ligand) act primarily as soluble trimers and serve to regulate the steady-state numbers of nearly all B-cell compartments. This homeostatic regulation is accomplished through the regulation of B-cell production rates, selection thresholds, and lifespan. Differential expression of the three BLyS receptors during differentiation and activation provides related yet distinct homeostatic niches for follicular, marginal zone, and memory B-cell subsets.
Collapse
Affiliation(s)
- Michael P Cancro
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6082, USA.
| |
Collapse
|
98
|
Salaün P, Boulben S, Mulner-Lorillon O, Bellé R, Sonenberg N, Morales J, Cormier P. Embryonic-stage-dependent changes in the level of eIF4E-binding proteins during early development of sea urchin embryos. J Cell Sci 2005; 118:1385-94. [PMID: 15769855 DOI: 10.1242/jcs.01716] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The eukaryotic initiation factor 4E (eIF4E)-binding proteins (4E-BPs) inhibit translation initiation by binding eIF4E and preventing recruitment of the translation machinery to mRNA. We have previously shown that fertilization of sea urchin eggs triggers eIF4E-4E-BP complex dissociation and 4E-BP degradation. Here, we show that microinjection of eIF4E-binding motif peptide into unfertilized eggs delays the onset of the first mitosis triggered by fertilization, demonstrating that dissociation of the eIF4E-4E-BP complex is functionally important for the first mitotic division in sea urchin embryos. We also show by gel filtration analyses that eIF4E is present in unfertilized eggs as an 80 kDa molecular mass complex containing 4E-BP and a new 4E-BP of 40 kDa. Fertilization triggers the dissociation of eIF4E from these two 4E-BPs and triggers the rapid recruitment of eIF4E into a high-molecular-mass complex. Release of eIF4E from the two 4E-BPs is correlated with a decrease in the total level of both 4E-BPs following fertilization. Abundance of the two 4E-BPs has been monitored during embryonic development. The level of the two proteins remains very low during the rapid cleavage stage of early development and increases 8 hours after fertilization. These results demonstrate that these two 4E-BPs are down- and upregulated during the embryonic development of sea urchins. Consequently, these data suggest that eIF4E availability to other partners represents an important determinant of the early development of sea urchin embryos.
Collapse
Affiliation(s)
- Patrick Salaün
- Station Biologique de Roscoff, Cycle Cellulaire et Développement, Unité Mer et Santé (UMR 7150), Université Pierre et Marie Curie (EI 37), Centre National de la Recherche Scientifique (CNRS), BP 74, 29682 Roscoff Cedex, France
| | | | | | | | | | | | | |
Collapse
|
99
|
Qiuping Z, Jie X, Youxin J, Qun W, Wei J, Chun L, Jin W, Yan L, Chunsong H, Mingzhen Y, Qingping G, Qun L, Kejian Z, Zhimin S, Junyan L, Jinquan T. Selectively frequent expression of CXCR5 enhances resistance to apoptosis in CD8(+)CD34(+) T cells from patients with T-cell-lineage acute lymphocytic leukemia. Oncogene 2005; 24:573-84. [PMID: 15580304 DOI: 10.1038/sj.onc.1208184] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We investigated CD4(+)CD34(+), CD8(+)CD34(+), CD4(+)CD34(-), and CD8(+)CD34(-) T cells from cord blood and from typical patients with T-cell-lineage acute lymphocytic leukemia and T-cell-lineage chronic lymphocytic leukemia in terms of expression and functions of CXCR5/CXCL13. We found that CXCR5 was selectively frequently expressed on T-cell-lineage acute (chronic) lymphocytic leukemia (T-ALL) CD8(+)CD34(+) T cells, but not on T-ALL CD4(+)CD34(+), CD4(+)CD34(-), and CD8(+)CD34(-) T cells. CXCR5 was rarely expressed on all types of CD34(+) and CD34(-) CB or T-CLL T cells. CXCL13/B cells attracting chemokine 1 induced significant resistance to TNF-alpha-mediated apoptosis in T-ALL CD8(+)CD34(+) T cells, instead of induction of chemotactic and adhesive responsiveness. A proliferation-inducing ligand expression in T-ALL CD8(+)CD34(+) T cells was upregulated by CXCL13/BCA-1 (B-cell attracting chemokine 1). The CXCR5/CXCL13 pair by means of activation of APRIL (A proliferation-inducing ligand) induced resistance to apoptosis in T-ALL CD8(+)CD34(+) T cells in livin-dependent manner. In this process, cell-cell contact in culture was necessary. Based on our findings, we suggested that there were differential functions of CXCR5/CXCL13 in distinct types of cells. Normal lymphocytes, especially naive B and T cells, utilized CXCR5/CXCL13 for migration, homing, maturation, and cell homeostasis, as well as secondary lymphoid tissue organogenesis. Meanwhile, certain malignant cells took advantages of CXCR5/CXCL13 for infiltration, resistance to apoptosis, and inappropriate proliferation.
Collapse
Affiliation(s)
- Zhang Qiuping
- Department of Immunology, Institute of Allergy and Immune-related Diseases, Centre for Medical Research, Wuhan University School of Medicine, Wuhan University, Dong Hu Road 115, Wuchang, Wuhan 430071, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Ho DH, Vu H, Brown SAN, Donohue PJ, Hanscom HN, Winkles JA. Soluble tumor necrosis factor-like weak inducer of apoptosis overexpression in HEK293 cells promotes tumor growth and angiogenesis in athymic nude mice. Cancer Res 2005; 64:8968-72. [PMID: 15604260 DOI: 10.1158/0008-5472.can-04-1879] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) is a member of the tumor necrosis factor superfamily of structurally related cytokines. TWEAK acts on responsive cells via binding to a cell surface receptor named Fn14. Recent studies have demonstrated that TWEAK can stimulate numerous cellular responses including cell proliferation, migration, and proinflammatory molecule production. It has also been reported that TWEAK can stimulate blood vessel formation in the rat cornea angiogenesis assay, but it is presently unknown whether this cytokine could play a role in the pathological angiogenesis associated with human diseases such as cancer, rheumatoid arthritis, and diabetic retinopathy. In the present study we investigated whether TWEAK was expressed in human tumors and whether it could promote tumor growth and angiogenesis in vivo. TWEAK mRNA expression was detected in many tumor types by cDNA array hybridization analysis, and TWEAK protein expression was confirmed in human colon cancer tissue by immunohistochemistry. As an initial approach to address whether TWEAK might act as a tumor angiogenesis factor, we established several human embryonic kidney cell lines that constitutively secrete a soluble TWEAK protein and examined their growth properties in vitro and in vivo. We found that although TWEAK-overexpressing cells do not have a growth advantage in vitro, they form larger and more highly vascularized tumors in athymic mice when compared with control, vector-transfected cells. This result suggests that the TWEAK-Fn14 signaling system may be a potential regulator of human tumorigenesis.
Collapse
Affiliation(s)
- David H Ho
- Department of Surgery and Physiology and the University of Maryland Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | |
Collapse
|