Decoy Receptor 3 Ameliorates an Autoimmune Crescentic Glomerulonephritis Model in Mice

Total Page:16

File Type:pdf, Size:1020Kb

Decoy Receptor 3 Ameliorates an Autoimmune Crescentic Glomerulonephritis Model in Mice JASN Express. Published on August 8, 2007 as doi: 10.1681/ASN.2006111242 BASIC RESEARCH www.jasn.org Decoy Receptor 3 Ameliorates an Autoimmune Crescentic Glomerulonephritis Model in Mice ʈ Shuk-Man Ka,* Huey-Kang Sytwu,†‡ Deh-Ming Chang,§ Shie-Liang Hsieh, Pei-Yi Tsai,† and Ann Chen* *Department of Pathology, §Division of Rheumatology/Immunology & Allergy, Department of Medicine, Tri-Service General Hospital, †Graduate Institute of Medical Sciences, ‡Department of Microbiology and Immunology, National ʈ Defense Medical Center, and Institute and Department of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan, Republic of China ABSTRACT Autoimmune crescentic glomerulonephritis (ACGN) is a variant of crescentic glomerulonephritis. The outcome of treatment of crescentic glomerulonephritis is poor. Binding of decoy receptor 3 (DCR3) to its ligand is capable of downregulating the alloresponsiveness of T cells. DCR3 has also been shown to benefit an experimental autoimmune model of diabetes. This study tested the hypothesis that a potential immune regulator, DCR3, could prevent the evolution of ACGN. With the use of an established ACGN model in mice, mice were treated with 100 ␮g/10 g body wt human DCR3 by hydrodynamics- based gene delivery at 14-d intervals. The results showed that the gene therapy resulted in (1) suppression of T and B cell activation and T cell proliferation; (2) a reduction in serum levels of proinflammatory cytokines; (3) improvement of proteinuria and renal dysfunction; (4) prevention of glomerular crescent formation, renal interstitial inflammation, and glomerulosclerosis; (5) a reduction in serum levels of autoantibodies and glomerular immune deposits; (6) inhibition of apoptosis in the spleen and kidney; (7) prevention of T cell and macrophage infiltration of the kidney; and (8) suppression of fibrosis-related gene expression in the kidney compared with empty vector–treated (disease control) ACGN mice. On the basis of these findings, it is proposed that human DCR3 exerts its preventive and protective effects on ACGN through modulation of T cell activation/proliferation, B cell activation, protection against apoptosis, and suppression of mononuclear leukocyte infiltration in the kidney. J Am Soc Nephrol 18: 2473–2485, 2007. doi: 10.1681/ASN.2006111242 Systemic lupus erythematosus (SLE) is a typical au- (ACGN) is an extremely progressive form of lupus toimmune disease involving multiple organs. Al- nephritis and is classified as a type of crescentic though its cause is not entirely clear, the basic ab- glomerulonephritis11,12 in which widespread normality seems to be a failure of mechanisms that glomerular crescents, consisting of a mixture of maintain self-tolerance, resulting in the production parietal epithelial cells, monocytes/macrophages, of diverse autoantibodies, especially antinuclear an- tibodies,1 T cell abnormalities,2,3 and apoptosis.4–6 In patients and mouse models, pathogenic T cells Received November 15, 2006. Accepted May 24, 2007. recognize self-antigens and drive B cell hyperactiv- Published online ahead of print. Publication date available at ity, confirming their central role in the pathogenesis www.jasn.org. 3,7–9 of SLE. Lupus nephritis is a major renal injury Correspondence: Dr. Ann Chen, Department of Pathology, Tri- in SLE and is characterized by immune complex Service General Hospital, National Defense Medical Center, No. deposition in the glomerular and renal tubular and 325, Sec. 2, Cheng-Gung Road, Taipei, Taiwan, ROC, Phone: ϩ886-2-8792-7008; Fax: ϩ886-2-8792-7009; E-mail: doc31717@ 10 peritubular capillary basement membranes. Clin- ndmctsgh.edu.tw ically, autoimmune crescentic glomerulonephritis Copyright © 2007 by the American Society of Nephrology J Am Soc Nephrol 18: 2473–2485, 2007 ISSN : 1046-6673/1809-2473 2473 BASIC RESEARCH www.jasn.org and lymphocytes, are formed.13,14 We have shown that a ner and significantly reduces the severity of insulitis in an au- murine chronic graft-versus-host disease, induced in toimmune diabetes model. In addition, in vivo administration C57BL/6 ϫ DBA/2J F1 hybrid mice by giving DBA/2J donor of hDCR3 ameliorates allograft rejection.17 The effects of lymphocytes, can progress to ACGN, with extensive, char- hDCR3 on immune regulation should be explored for its pos- acteristic glomerular crescent formation (up to 80% of glo- sible therapeutic use in controlling undesirable immune re- meruli examined), sclerosis, and intense interstitial inflam- sponses. Clinically, DCR3 gene expression has been detected in mation.15 Although recovery of renal functions in patients peripheral blood mononuclear leukocytes derived from pa- with crescentic glomerulonephritis can occur after early in- tients with SLE,24 although its clinical significance remains to tensive plasma exchange and/or treatment with steroids and be determined. cytotoxic agents, patients eventually require long-term di- In this study, we tested the hypothesis that in vivo over- alysis or transplantation.11 expression of hDCR3 would prevent crescentic formation of Decoy receptor 3 (DCR3) lacks the transmembrane do- an experimental ACGN. Using hydrodynamics-based gene main of the conventional TNF receptor and is a secreted delivery, we demonstrated that hDCR3 gene therapy is an protein.16,17 DCR3 can interact with Fas ligand (FasL),16 effective therapeutic approach in the ACGN model. Regu- LIGHT (homologous to lymphotoxins, shows inducible ex- lation of both T cell function and apoptosis in lymphoid pression and competes with HSV glycoprotein D for herpes organs and in the kidney seems to be the major factor re- virus entry mediator, a receptor expressed by T lympho- sponsible for its favorable effects in this autoimmune kid- cytes),18 and TNF-like molecule 1A.19 Binding of DCR3 to ney disease model. LIGHT also downregulates the alloresponsiveness of T cells.17,20 By helping tumor cells avoid immune attack through lymphocyte infiltration and FasL/LIGHT-medi- RESULTS ated apoptosis, increased DCR3 expression might benefit their growth.16,18 DCR3 exerts another regulatory function Serum Levels of hDCR3 Protein in Treated ACGN by directly modulating the differentiation and function of Mice macrophages and dendritic cells.21,22 Based on the hDCR3 expression (Figure 1, methods as de- Recently, Sung et al.23 reported that transgenic human scribed in Concise Methods), we delivered the hDCR3 plasmid DCR3 (hDCR3) protects mice from autoimmune and cyclo- to the mice at 14-d intervals. After administrations of hDCR3 phosphamide-induced diabetes in a dosage-dependent man- plasmid or empty vector, ELISA tests showed high serum levels Figure 1. Serum levels and hepatic expression of human decoy receptor 3 (hDCR3) protein. (A) Serum hDCR3 levels in normal control mice after a single injection of hDCR3 plasmid or empty vector measured by ELISA. Data are means Ϯ SEM for groups of Figure 2. Serum levels of hDCR3 in treated autoimmune crescen- 10 mice. The arrow indicates the time of gene delivery with tic glomerulonephritis (ACGN) mice. Serum levels at week 3 (A) hDCR3 or empty vector. (B) hDCR3 protein expression in the liver and week 9 (B) in ACGN mice after administration of hDCR3 or of normal control mice on day 2 after a single injection of hDCR3 empty vector. Data are means Ϯ SEM for groups of 10 mice. #Not plasmid or empty vector. Magnification, ϫ400. detectable. 2474 Journal of the American Society of Nephrology J Am Soc Nephrol 18: 2473–2485, 2007 www.jasn.org BASIC RESEARCH of hDCR3 protein in the hDCR3-treated ACGN mice at 3 wk empty vector–treated ACGN mice at 3 wk (Figure 3, A and C) (Figure 2A) and 9 wk (Figure 2B), respectively, after ACGN or 9 wk (Figure 3, B and D) after ACGN induction, compared induction but not in the empty vector–treated ACGN mice or with normal control mice (both P Ͻ 0.005). However, this normal control mice. effect was significantly suppressed by administration of hDCR3 plasmid to ACGN mice compared with empty vec- DCR3 Suppresses T/B Cell Activities tor–treated ACGN mice (P Ͻ 0.05 at 3 wk; P Ͻ 0.005 at 9 Inhibition of T/B Cell Activation. wk). Moreover, hDCR3-treated ACGN mice showed a sig- ϩ ϩ Abnormal T and B cell cooperation can cause a graft-versus- nificantly lower percentage of CD4 CD69 cells (activated host reaction,9,25 the basic mechanism for the induction of the T helper cells) than that of empty vector–treated ACGN ACGN model. DCR3 has been reported to inhibit T cell pro- mice at week 3 (4.0 32 Ϯ 1.0 versus 8.1 Ϯ 1.5%; P Ͻ 0.05) and liferation and lymphokine secretion.17,19,21 We tested whether week 9 (9.5 Ϯ 0.5 versus 18.2 Ϯ 2.9%; P Ͻ 0.05), respec- ϩ ϩ hDCR3 could ameliorate the development of the ACGN model tively. Conversely, the percentage of CD19 CD69 cells by negatively regulating T cell and/or B cell activation by per- (activated B cells) was significantly decreased in hDCR3- forming flow cytometry of splenocytes. treated ACGN mice to the levels seen in normal control As shown in Figure 3, the percentage of CD3ϩCD69ϩ cells mice at week 9 (P Ͻ 0.005; Figure 3, B and D), although (activated T cells) in the spleen was significantly increased in there was no significant difference in the percentage of ϩ Figure 3. Flow cytometry for T and B cells in the spleen. Immunofluorescence dot-plot pattern of the CD69 activation marker on CD3 ϩ ϩ ϩ ϩ ϩ T cells or CD19 B cells. (A and C) Week 3. (B and D) Week 9. (C and D) Percentage of CD3 CD69 T cells and CD19 CD69 . Each bar represents the means Ϯ SEM for groups of 10 mice. *P Ͻ 0.05; ***P Ͻ 0.005; NS, no significant difference. J Am Soc Nephrol 18: 2473–2485, 2007 Decoy Receptor 3 Ameliorates Lupus Nephritis 2475 BASIC RESEARCH www.jasn.org CD19ϩCD69ϩ cells between empty vector–treated ACGN and hDCR3-treated ACGN mice (Figure 3, A and C) at week 3 compared with empty vector–treated ACGN mice.
Recommended publications
  • Cytokine Nomenclature
    RayBiotech, Inc. The protein array pioneer company Cytokine Nomenclature Cytokine Name Official Full Name Genbank Related Names Symbol 4-1BB TNFRSF Tumor necrosis factor NP_001552 CD137, ILA, 4-1BB ligand receptor 9 receptor superfamily .2. member 9 6Ckine CCL21 6-Cysteine Chemokine NM_002989 Small-inducible cytokine A21, Beta chemokine exodus-2, Secondary lymphoid-tissue chemokine, SLC, SCYA21 ACE ACE Angiotensin-converting NP_000780 CD143, DCP, DCP1 enzyme .1. NP_690043 .1. ACE-2 ACE2 Angiotensin-converting NP_068576 ACE-related carboxypeptidase, enzyme 2 .1 Angiotensin-converting enzyme homolog ACTH ACTH Adrenocorticotropic NP_000930 POMC, Pro-opiomelanocortin, hormone .1. Corticotropin-lipotropin, NPP, NP_001030 Melanotropin gamma, Gamma- 333.1 MSH, Potential peptide, Corticotropin, Melanotropin alpha, Alpha-MSH, Corticotropin-like intermediary peptide, CLIP, Lipotropin beta, Beta-LPH, Lipotropin gamma, Gamma-LPH, Melanotropin beta, Beta-MSH, Beta-endorphin, Met-enkephalin ACTHR ACTHR Adrenocorticotropic NP_000520 Melanocortin receptor 2, MC2-R hormone receptor .1 Activin A INHBA Activin A NM_002192 Activin beta-A chain, Erythroid differentiation protein, EDF, INHBA Activin B INHBB Activin B NM_002193 Inhibin beta B chain, Activin beta-B chain Activin C INHBC Activin C NM005538 Inhibin, beta C Activin RIA ACVR1 Activin receptor type-1 NM_001105 Activin receptor type I, ACTR-I, Serine/threonine-protein kinase receptor R1, SKR1, Activin receptor-like kinase 2, ALK-2, TGF-B superfamily receptor type I, TSR-I, ACVRLK2 Activin RIB ACVR1B
    [Show full text]
  • TNF Decoy Receptors Encoded by Poxviruses
    pathogens Review TNF Decoy Receptors Encoded by Poxviruses Francisco Javier Alvarez-de Miranda † , Isabel Alonso-Sánchez † , Antonio Alcamí and Bruno Hernaez * Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Campus de Cantoblanco, Universidad Autónoma de Madrid, Nicolás Cabrera 1, 28049 Madrid, Spain; [email protected] (F.J.A.-d.M.); [email protected] (I.A.-S.); [email protected] (A.A.) * Correspondence: [email protected]; Tel.: +34-911-196-4590 † These authors contributed equally. Abstract: Tumour necrosis factor (TNF) is an inflammatory cytokine produced in response to viral infections that promotes the recruitment and activation of leukocytes to sites of infection. This TNF- based host response is essential to limit virus spreading, thus poxviruses have evolutionarily adopted diverse molecular mechanisms to counteract TNF antiviral action. These include the expression of poxvirus-encoded soluble receptors or proteins able to bind and neutralize TNF and other members of the TNF ligand superfamily, acting as decoy receptors. This article reviews in detail the various TNF decoy receptors identified to date in the genomes from different poxvirus species, with a special focus on their impact on poxvirus pathogenesis and their potential use as therapeutic molecules. Keywords: poxvirus; immune evasion; tumour necrosis factor; tumour necrosis factor receptors; lymphotoxin; inflammation; cytokines; secreted decoy receptors; vaccinia virus; ectromelia virus; cowpox virus Citation: Alvarez-de Miranda, F.J.; Alonso-Sánchez, I.; Alcamí, A.; 1. TNF Biology Hernaez, B. TNF Decoy Receptors TNF is a potent pro-inflammatory cytokine with a broad range of biological effects, Encoded by Poxviruses. Pathogens ranging from the activation of inflammatory gene programs to cell differentiation or 2021, 10, 1065.
    [Show full text]
  • CD95 Ligand - Death Factor and Costimulatory Molecule?
    Cell Death and Differentiation (2003) 10, 1215–1225 & 2003 Nature Publishing Group All rights reserved 1350-9047/03 $25.00 www.nature.com/cdd Review CD95 ligand - death factor and costimulatory molecule? O Janssen*,1, J Qian1, A Linkermann1 and D Kabelitz1 Tissue and Cellular Expression of CD95L 1 Institute for Immunology, Medical Center Schleswig-Holstein, Campus Kiel, Michaelisstrasse 5, D-24105 Kiel, Germany The CD95 ligand (CD95L, Apo-1L, FasL, CD178) is a 281- * Corresponding author: O Janssen. Tel: þ 49-431-5973377; Fax: þ 49-431- amino-acid-containing type II transmembrane protein of the 5973335; E-mail: [email protected] TNF family of death factors (Figure 1).1 Its death-inducing function is best documented in the context of activation- Received 24.4.03; revised 12.6.03; accepted 20.6.03; published online 1 August 2003 induced cell death (AICD) in T cells.2 CD95L is expressed as a Edited by T Ferguson death factor in cytotoxic T lymphocytes (CTL) to kill virally infected or transformed target cells and in natural killer (NK) cells, where it is upregulated by CD16 engagement and 3 Abstract cytokines including IL-2 and IL-12. Similarly, high levels of intracellular CD95L have been detected in monocytic cells The CD95 ligand is involved as a death factor in the with an inducible release upon activation.4 Under physiologi- regulation of activation-induced cell death, establishment cal conditions, CD95L is implicated in the control of erythroid of immune privilege and tumor cell survival. In addition, differentiation,5 angiogenesis in the eye6 and skin home- 7 CD95L may serve as a costimulatory molecule for T-cell ostasis.
    [Show full text]
  • Decoy Strategies: the Structure of TL1A:Dcr3 Complex
    Structure Article Decoy Strategies: The Structure of TL1A:DcR3 Complex Chenyang Zhan,1 Yury Patskovsky,1 Qingrong Yan,2 Zhenhong Li,1 Udupi Ramagopal,1 Huiyong Cheng,1 Michael Brenowitz,1 Xiao Hui,3 Stanley G. Nathenson,2,4 and Steven C. Almo1,5,* 1Department of Biochemistry 2Department of Cell Biology 3Department of Developmental and Molecular Biology 4Department of Microbiology and Immunology 5Department of Physiology and Biophysics Albert Einstein College of Medicine, Bronx, NY 10461, USA *Correspondence: [email protected] DOI 10.1016/j.str.2010.12.004 SUMMARY domains (CRDs), which bind the interprotomer grooves formed between adjacent ligand monomers. This mode of interaction Decoy Receptor 3 (DcR3), a secreted member of the results in ligand:receptor assemblies with 3:3 stoichiometries Tumor Necrosis Factor (TNF) receptor superfamily, that span two interacting cells. The ligand-mediated clustering neutralizes three different TNF ligands: FasL, LIGHT, of TNFRs directs the recruitment of signaling adaptor proteins, and TL1A. Each of these ligands engages unique such as TNF receptor associated factors (TRAFs) or death signaling receptors which direct distinct and critical domain adaptor proteins, and initiates diverse downstream immune responses. We report the crystal structures signaling pathways. Some TNFRs are also produced as soluble forms due to alter- of the unliganded DcR3 ectodomain and its complex nate mRNA splicing or selective proteolytic cleavage (Cheng with TL1A, as well as complementary mutagenesis et al., 1994; Locksley et al., 2001; Van Zee et al., 1992). The met- and biochemical studies. These analyses demon- alloprotease-mediated cleavage of two TNF-a receptors (TNFR1 strate that DcR3 interacts with invariant backbone and TNFR2) has been reported to attenuate proinflammatory and side-chain atoms in the membrane-proximal TNF-a-associated signaling, possibly by reducing the amount half of TL1A which supports recognition of its three of cell surface receptors and by the sequestration/neutralization distinct TNF ligands.
    [Show full text]
  • Decoy Receptor 3 (Dcr3) Induces Osteoclast Formation from Monocyte/Macrophage Lineage Precursor Cells
    Cell Death and Differentiation (2004) 11, S97–S107 & 2004 Nature Publishing Group All rights reserved 1350-9047/04 $30.00 www.nature.com/cdd Decoy receptor 3 (DcR3) induces osteoclast formation from monocyte/macrophage lineage precursor cells CR Yang1, JH Wang2, SL Hsieh3, SM Wang4, TL Hsu3 and lineage. 1,2 Osteoclasts and macrophages are known to have WW Lin*,1 several morphological, cytochemical, and functional charac- teristics in common, and are derived from the same committed 1 Department of Pharmacology, College of Medicine, National Taiwan population of hematopoietic precursors.3 In vitro studies have University, Taipei, Taiwan shown that osteoclasts in both mouse and humans may form 2 Department of Orthopedic Surgery, National Taiwan University Hospital, directly from precursor cell populations of monocytes and Taipei, Taiwan macrophages.4 3 Department of Microbiology and Immunology, and Immunology Research Center, National Yang-Ming University, Taipei, Taiwan To date, at least two key molecules that are essential and 4 Department of Anatomy and Cell Biology, College of Medicine, National sufficient to the promotion of osteoclastogenesis have been Taiwan University, Taipei, Taiwan identified, that is, macrophage colony-stimulating factor (M- * Corresponding author: WW Lin, Department of Pharmacology, College of CSF) and the receptor for activation of NF-kB ligand (RANKL).5 Medicine, National Taiwan University, Taipei, Taiwan. Tel: þ 886-2-23123456 M-CSF, which is imperative for macrophage maturation, binds ext. 8315; Fax: þ 886-2-23915297; E-mail: [email protected] to its receptor on early osteoclast precursors, thereby providing signals required for their survival, proliferation, and differentia- Received 28.10.03; revised 03.12.03; accepted 29.12.03; published online 27.2.04 4,6 Edited by M Piacentini tion.
    [Show full text]
  • The Vital Osteoclast: How Is It Regulated?
    Cell Death and Differentiation (2004) 11, S5–S7 & 2004 Nature Publishing Group All rights reserved 1350-9047/04 $30.00 www.nature.com/cdd Editorial The vital osteoclast: how is it regulated? D Aeschlimann*,1 and BAJ Evans2 exogenous cholesterol/lipoprotein levels. In the following, these findings are briefly discussed in their respective 1 Matrix Biology & Tissue Repair Research Unit, Dental School, University of contexts. Wales College of Medicine, Heath Park, Cardiff, CF14 4XY, UK In addition to M-CSF and RANKL, pro-inflammatory 2 Department of Child Health, University of Wales College of Medicine, Heath cytokines – TNF-a in particular – have been shown to have Park, Cardiff, CF14 4XY, UK crucial roles in osteoclast differentiation. Oestrogen defi- * Corresponding author: D Aeschlimann; E-mail: [email protected] ciency induces bone loss as a result of alterations in cytokine expression that either directly or indirectly modulate osteo- Cell Death and Differentiation (2004) 11, S5–S7. clasts and their precursors, and thereby increase resorption. doi:10.1038/sj.cdd.4401470 TNF receptor 1 (TNFR1)-deficient mice are not responsive to oestrogen depletion, and TNF-a, acting through TNFR1, Bone is a rigid but dynamic organ. Once formed, it is augments RANKL-induced osteoclastogenesis.5,6 It has also continually broken down and reformed by the co-ordinated been shown that high concentrations of TNF-a promote in vitro actions of osteoclasts (that mediate resorption) and osteo- osteoclast formation independently of RANKL, through NF-kB blasts (that mediate formation) on trabecular bone surfaces and JNK activation.7 However, the complete lack of osteo- and in the Haversian systems of cortical bone.
    [Show full text]
  • Role of Tgfβ3-Smads-Sp1 Axis in Dcr3-Mediated Immune
    Zhu et al. Oncogenesis (2019) 8:43 https://doi.org/10.1038/s41389-019-0152-0 Oncogenesis ARTICLE Open Access Role of TGFβ3-Smads-Sp1 axis in DcR3-mediated immune escape of hepatocellular carcinoma Hui-fang Zhu1,2, Yan-ping Liu1, Ding-li Liu3,Yi-danMa1,Zhi-yanHu1,Xiao-yanWang1, Chuan-sha Gu1,2, Yan Zhong1, Ting Long1,He-pingKan4 and Zu-guo Li1,5,6 Abstract Hepatocellular carcinoma (HCC) is a leading cause of tumour-associated mortality worldwide, but no significant improvement in treating HCC has been reported with currently available systemic therapies. Immunotherapy represents a new frontier in tumour therapy. Therefore, the immunobiology of hepatocarcinoma has been under intensive investigation. Decoy receptor 3 (DcR3), a member of the tumour necrosis factor receptor (TNFR) superfamily, is an immune suppressor associated with tumourigenesis and cancer metastasis. However, little is known about the role of DcR3 in the immunobiology of hepatocarcinoma. In this study, we found that overexpression of DcR3 in HCC is mediated by the TGFβ3-Smad-Sp1 signalling pathway, which directly targets DcR3 promoter regions. Moreover, overexpression of DcR3 in HCC tissues is associated with tumour invasion and metastasis and significantly promotes the differentiation and secretion of Th2 and Treg cells while inhibiting the differentiation and secretion of Th1 cells. Conversely, knockdown of DcR3 expression in HCC significantly restored the immunity of CD4+ T cells. Inhibition of DcR3 expression may provide a novel immunotherapeutic approach to restoring immunity in HCC patients. 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; Introduction regulate the occurrence of HCC in such a micro- Hepatocellular carcinoma (HCC) is one of the most environment has not been elucidated to date.
    [Show full text]
  • Biolegend.Com
    Mechanisms of Cell Death TRAIL (TNFSF10) TNF-α Death Receptor 4 (TNFRSF10A/TRAIL-R1) Death Receptor 5 Zombie Dyes (TNFRSF10B/TRAIL-R2) Propidium Iodide (PI) BAT1, TIM-4 TNF RI (TNFRSF1A) 7-Amino-Actinomycin (7-AAD) MER TNF RII (TNFRSF1B) FAS-L GAS6 (TNFSF6/CD178) TRAIL (TNFSF10) Apoptotic Cell Death Domain Zombie Dyes Phosphatidylserine K63 Ubiquitin NH2 Removal ICAM3? ROCK1 NH CD14 2 Eat-Me Signals FAS Death Inducing Cytoskeletal Rearrangement, (TNFRSF6/CD95) Signaling Complex (DISC) TRADD Cytoskeletal Rearrangement, TRADD Decoy Receptor 2 FADD (TNFRSF10D/TRAIL-R4) Actomysin Contraction Engulfment RIP1 TWEAK RIP1 oxLDL (TNFSF12) FADD CIAP1/2 K63 Ubiquitination Blebbing CD36 Death Receptor 3 TWEAK (TNFSF12) PI FADD (TNFRSF25, APO-3) 7-AAD TRAF1 FADD Procaspase 8,10 TRAF 3 Phagocyte FLIP PANX1 Macrophage Monocyte Neutrophil Dendritic Cell Fibroblast Mast Cell Procaspase 8,10 NF-kB TWEAK-R (TNFRSF12A/Fn14) Find-Me Signals Lysophosphocholine C Caspase 8,10 TRAF5 TRAF2 Sphingosine-1-Phosphate G2A? Nucleotides A Decoy TRAIL Receptor R1 (TNFRSF23) Bid Cell Survival ATP, UTP Decoy TRAIL Receptor R2 (TNFRSF22) Sphingosine-1 TRADD Phosphate Receptor Decoy Receptor 1 (TNFRSF10C/TRAIL-R3) Procaspase 3 Proliferation RIP1 G P2y2 t-Bid Bcl-2 T Chemotaxis, Caspase 3 Bcl-2-xL, MCL-1 ? ICAD RIP1 Engulfment Degradation Bax, Bak Oligomerization TRADD Death Receptor 6 Extracellular ATP Bacterial pore-forming toxins TRAIL (TNFSF10) ICAD (TNFRSF21) Monosodium urate crystals Cholesterol crystals Death Receptor DNA Fragmentation Cholera toxin B, Mitochondria
    [Show full text]
  • Dcr3/Fc Chimera, Human (D2441)
    DcR3/Fc Chimera, human recombinant, expressed in Sf 21 cells Catalog Number D2441 Storage Temperature -20 °C Product Description Preparation Instructions Decoy Receptor 3 (DcR3) is produced from a DNA Reconstitute the contents of the vial using sterile sequence encoding the signal peptide from human phosphate buffered saline containing at least 0.1% CD33 joined with amino acid residues 24-300 of human human serum albumin or bovine serum albumin. DcR31 fused to the Fc region of human IgG1 by a Prepare a stock solution of no less than 10 mg/mL. polypeptide linker. The reduced human DcR3/Fc monomer has a calculated molecular mass of 57 kDa. Precautions and Disclaimer Due to glycosylation, the recombinant protein migrates This product is for R&D use only, not for drug, as a 60-65 kDa protein in SDS-PAGE under reducing household, or other uses. Please consult the Material conditions. Based on N-terminal sequencing, the Safety Data Sheet for information regarding hazards protein has Val24 at the N-terminus. DcR3 shares and safe handling practices. amino acid sequence homology with osteoprotegerin (31%), TNFRII (29%), and Fas (17%). Storage/Stability Store at -20 °C. Upon reconstitution, the product may Apoptosis, or programmed cell death, is induced in cells be stored at 2-8 °C for up to one month. For extended by a group of death domain-containing receptors storage, freeze in working aliquots at -20 °C. Repeated including TNFR1, Fas, DR3, DR4, and DR5. Binding of freezing and thawing, or storage in a “frost-free” ligand to these receptors sends signals that activate freezers, is not recommended.
    [Show full text]
  • Licensed Human Natural Killer Cells Aid Dendritic Cell Maturation Via TNFSF14/LIGHT
    Licensed human natural killer cells aid dendritic cell maturation via TNFSF14/LIGHT Tim D. Holmesa,1, Erica B. Wilsona, Emma V. I. Blacka, Andrew V. Benesta,2, Candida Vazb, Betty Tanb, Vivek M. Tanavdeb,c, and Graham P. Cooka,3 aLeeds Institute of Cancer and Pathology, University of Leeds School of Medicine, St. James’s University Hospital, Leeds LS9 7TF, United Kingdom; and bBioinformatics Institute and cInstitute of Medical Biology, A*STAR (Agency for Science, Technology and Research), Biopolis, Singapore 138671 Edited by Wayne M. Yokoyama, Washington University School of Medicine, St. Louis, MO, and approved November 19, 2014 (received for review June 13, 2014) Interactions between natural killer (NK) cells and dendritic cells adaptive immunity. This potent cytotoxicity and proinflammatory (DCs) aid DC maturation and promote T-cell responses. Here, we activity must be strictly controlled to minimize damage to healthy have analyzed the response of human NK cells to tumor cells, and tissue. Functional competency of unstimulated NK cells is – we identify a pathway by which NK DC interactions occur. Gene achieved via a process termed “licensing” or “education” (11– expression profiling of tumor-responsive NK cells identified the very 14). Licensing ensures that only those NK cells expressing in- rapid induction of TNF superfamily member 14 [TNFSF14; also hibitory receptors for self-MHC class I can respond to target cells known as homologous to lymphotoxins, exhibits inducible expres- sion, and competes with HSV glycoprotein D for HVEM, a receptor and NK cells that lack inhibitory receptors for self-MHC class I expressed by T lymphocytes (LIGHT)], a cytokine implicated in the molecules are rendered hyporesponsive, preventing them from enhancement of antitumor responses.
    [Show full text]
  • Proteolysis Generated by Alternate Splicing Or Chromosome 19P13.3
    Genomic Characterization of LIGHT Reveals Linkage to an Immune Response Locus on Chromosome 19p13.3 and Distinct Isoforms Generated by Alternate Splicing or This information is current as Proteolysis of September 23, 2021. Steve W. Granger, Kris D. Butrovich, Pantea Houshmand, Wilson R. Edwards and Carl F. Ware J Immunol 2001; 167:5122-5128; ; doi: 10.4049/jimmunol.167.9.5122 Downloaded from http://www.jimmunol.org/content/167/9/5122 References This article cites 34 articles, 14 of which you can access for free at: http://www.jimmunol.org/ http://www.jimmunol.org/content/167/9/5122.full#ref-list-1 Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists by guest on September 23, 2021 • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2001 by The American Association of Immunologists All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Genomic Characterization of LIGHT Reveals Linkage to an Immune Response Locus on Chromosome 19p13.3 and Distinct Isoforms Generated by Alternate Splicing or Proteolysis1,2 Steve W. Granger, Kris D.
    [Show full text]
  • Cell Structure & Function
    Cell Structure & Function Antibodies and Reagents BioLegend is ISO 13485:2016 Certified Toll-Free Tel: (US & Canada): 1.877.BIOLEGEND (246.5343) Tel: 858.768.5800 biolegend.com 02-0012-03 World-Class Quality | Superior Customer Support | Outstanding Value Table of Contents Introduction ....................................................................................................................................................................................3 Cell Biology Antibody Validation .............................................................................................................................................4 Cell Structure/ Organelles ..........................................................................................................................................................8 Cell Development and Differentiation ................................................................................................................................10 Growth Factors and Receptors ...............................................................................................................................................12 Cell Proliferation, Growth, and Viability...............................................................................................................................14 Cell Cycle ........................................................................................................................................................................................16 Cell Signaling ................................................................................................................................................................................18
    [Show full text]