51
|
Tan T, Xia L. TRIM21 Aggravates Herpes Simplex Virus Epithelial Keratitis by Attenuating STING-IRF3-Mediated Type I Interferon Signaling. Front Microbiol 2020; 11:703. [PMID: 32373102 PMCID: PMC7176818 DOI: 10.3389/fmicb.2020.00703] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/26/2020] [Indexed: 12/30/2022] Open
Abstract
Herpes simplex virus-1 (HSV-1) is the leading cause of infectious blindness in the developed world. HSV-1 infection can occur anywhere in the eye, and the most common presentation is epithelial keratitis. In the HSV epithelial keratitis mice model, we detected the expression of TRIM21 and then investigated the clinical relationship between TRIM21 and HSV epithelial keratitis by silencing TRIM21. Through the clinical scores and histopathology examination, we found that TRIM21 can effectively reduce the severity of HSV epithelial keratitis. Furthermore, silencing TRIM21 significantly controlled the virus particle release at 1, 3, and 5 days post-HSV-1 infection. Notably, the production of IFN-β was enhanced, and the secretion of pro-inflammatory cytokines (IL-6 and TNF-a) was inhibited. Next, human corneal epithelial cells were pretreated with lentivirus or siRNA, respectively, so that TRIM21 expression was overexpressed or silenced. We focused on the regulation of STING-IRF3 and type I interferon signaling after infected with HSV-1. In conclusion, our results have identified that TRIM21 is abnormally high expressed in HSV epithelial keratitis. TRIM21 enhances the replication of HSV-1 in corneal epithelial cells via suppressing the production of type I IFN by inhibiting STING/IRF3 signaling. It also promotes the secretion of IL-6 and TNF-a, thereby aggravating the severity of HSV epithelial keratitis.
Collapse
Affiliation(s)
- Tianchang Tan
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Likun Xia
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
52
|
BNIP3L-Dependent Mitophagy Promotes HBx-Induced Cancer Stemness of Hepatocellular Carcinoma Cells via Glycolysis Metabolism Reprogramming. Cancers (Basel) 2020; 12:cancers12030655. [PMID: 32168902 PMCID: PMC7139741 DOI: 10.3390/cancers12030655] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 02/06/2023] Open
Abstract
Hepatitis B virus (HBV) is one of predisposing factors for hepatocellular carcinoma (HCC). The role of HBV x protein (HBx) in mediating the induction and maintenance of cancer stemness during HBV-related HCC attracts considerable attention, but the exact mechanism has not been clearly elucidated. Here, ABCG2-dependent stem-like side population (SP) cells, which are thought to be liver cancer stem cells (LCSCs), were present in HCC cells, and the fraction of this subset was increased in HBx-expressing HCC cells. In addition, glycolysis was upregulated in LCSCs and HBx-expressing HCC cells, and intervention of glycolysis attenuated cancer stem-like phenotypes. Mitochondria play an important role in the maintenance of energy homeostasis, BNIP3L-dependent mitophagy was also activated in LCSCs and HBx-expressing HCC cells, which triggered a metabolic shift toward glycolysis. In summary, we proposed a positive feedback loop, in which HBx induced BNIP3L-dependent mitophagy which upregulated glycolytic metabolism, increasing cancer stemness of HCC cells in vivo and in vitro. BNIP3L might be a potential therapeutic target for intervention of LCSCs-associated HCC. Anti-HBx, a monoclonal antibody targeting intracellular HBx, had the potential to delay the progression of HBV infection related-HCC.
Collapse
|
53
|
Gaston J, Maestrali N, Lalle G, Gagnaire M, Masiero A, Dumas B, Dabdoubi T, Radošević K, Berne PF. Intracellular delivery of therapeutic antibodies into specific cells using antibody-peptide fusions. Sci Rep 2019; 9:18688. [PMID: 31822703 PMCID: PMC6904672 DOI: 10.1038/s41598-019-55091-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 11/22/2019] [Indexed: 12/22/2022] Open
Abstract
Because of their favorable properties as macromolecular drugs, antibodies are a very successful therapeutic modality for interfering with disease-relevant targets in the extracellular space or at the cell membrane. However, a large number of diseases involve cytosolic targets and designing antibodies able to efficiently reach intracellular compartments would expand the antibody-tractable conditions. Here, we genetically fused cell penetrating peptides (CPPs) at various positions to an antibody targeting cancer cells, evaluated the developability features of the resulting antibody-peptide fusions and the ability of selected constructs to reach the cytosol. We first determined positions in the IgG structure that were permissive to CPP incorporation without destabilizing the antibody. Fusing CPPs to the C-terminus of the light chain and either before or after the hinge had the least effect on antibody developability features. These constructs were further evaluated for cell penetration efficiency. Two out of five tested CPPs significantly enhanced antibody penetration into the cytosol, in particular when fused before or after the hinge. Finally, we demonstrate that specific antibody binding to the cell surface target is necessary for efficient cell penetration of the CPP-antibody fusions. This study provides a solid basis for further exploration of therapeutic antibodies for intracellular targets.
Collapse
Affiliation(s)
- Julie Gaston
- Yubsis, 4 rue Pierre Fontaine, 91000, Evry, France
| | - Nicolas Maestrali
- Sanofi R&D, Biologics Research, 13 Quai Jules Guesde, 94400, Vitry-sur-Seine, France
| | - Guilhem Lalle
- Department of Immunology, Virology and Inflammation, UMR INSERM 1052, CNRS 5286, Centre Léon Bérard, Labex DEVweCAN, 693743, Lyon, France
| | - Marie Gagnaire
- Sanofi R&D, Biologics Research, 13 Quai Jules Guesde, 94400, Vitry-sur-Seine, France
| | - Alessandro Masiero
- Sanofi R&D, Biologics Research, 13 Quai Jules Guesde, 94400, Vitry-sur-Seine, France
| | - Bruno Dumas
- Sanofi R&D, Biologics Research, 13 Quai Jules Guesde, 94400, Vitry-sur-Seine, France
| | - Tarik Dabdoubi
- Sanofi R&D, Biologics Research, 13 Quai Jules Guesde, 94400, Vitry-sur-Seine, France
| | - Katarina Radošević
- Sanofi R&D, Biologics Research, 13 Quai Jules Guesde, 94400, Vitry-sur-Seine, France.
| | | |
Collapse
|
54
|
Xu J, Khan AR, Fu M, Wang R, Ji J, Zhai G. Cell-penetrating peptide: a means of breaking through the physiological barriers of different tissues and organs. J Control Release 2019; 309:106-124. [PMID: 31323244 DOI: 10.1016/j.jconrel.2019.07.020] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 07/15/2019] [Indexed: 12/24/2022]
Abstract
The selective infiltration of cell membranes and tissue barriers often blocks the entry of most active molecules. This natural defense mechanism prevents the invasion of exogenous substances and limits the therapeutic value of most available molecules. Therefore, it is particularly important to find appropriate ways of membrane translocation and therapeutic agent delivery to its target site. Cell penetrating peptides (CPPs) are a group of short peptides harnessed in this condition, possessing a significant capacity for membrane transduction and could be exploited to transfer various biologically active cargoes into the cells. Since their discovery, CPPs have been employed for delivery of a wide variety of therapeutic molecules to treat various disorders including cranial nerve involvement, ocular inflammation, myocardial ischemia, dermatosis and cancer. The promising results of CPPs-derived therapeutics in various tumor models demonstrated a potential and worthwhile scope of CPPs in chemotherapy. This review describes the detailed description of CPPs and CPPs-assisted molecular delivery against various tissues and organs disorders. An emphasis is focused on summarizing the novel insights and achievements of CPPs in surmounting the natural membrane barriers during the last 5 years.
Collapse
Affiliation(s)
- Jiangkang Xu
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Abdur Rauf Khan
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Manfei Fu
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Rujuan Wang
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Jianbo Ji
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Guangxi Zhai
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China.
| |
Collapse
|
55
|
Thura M, Al-Aidaroos AQ, Gupta A, Chee CE, Lee SC, Hui KM, Li J, Guan YK, Yong WP, So J, Chng WJ, Ng CH, Zhou J, Wang LZ, Yuen JSP, Ho HSS, Yi SM, Chiong E, Choo SP, Ngeow J, Ng MCH, Chua C, Yeo ESA, Tan IBH, Sng JXE, Tan NYZ, Thiery JP, Goh BC, Zeng Q. PRL3-zumab as an immunotherapy to inhibit tumors expressing PRL3 oncoprotein. Nat Commun 2019; 10:2484. [PMID: 31171773 PMCID: PMC6554295 DOI: 10.1038/s41467-019-10127-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 04/23/2019] [Indexed: 12/21/2022] Open
Abstract
Tumor-specific antibody drugs can serve as cancer therapy with minimal side effects. A humanized antibody, PRL3-zumab, specifically binds to an intracellular oncogenic phosphatase PRL3, which is frequently expressed in several cancers. Here we show that PRL3-zumab specifically inhibits PRL3+ cancer cells in vivo, but not in vitro. PRL3 antigens are detected on the cell surface and outer exosomal membranes, implying an 'inside-out' externalization of PRL3. PRL3-zumab binds to surface PRL3 in a manner consistent with that in classical antibody-dependent cell-mediated cytotoxicity or antibody-dependent cellular phagocytosis tumor elimination pathways, as PRL3-zumab requires an intact Fc region and host FcγII/III receptor engagement to recruit B cells, NK cells and macrophages to PRL3+ tumor microenvironments. PRL3 is overexpressed in 80.6% of 151 fresh-frozen tumor samples across 11 common cancers examined, but not in patient-matched normal tissues, thereby implicating PRL3 as a tumor-associated antigen. Targeting externalized PRL3 antigens with PRL3-zumab may represent a feasible approach for anti-tumor immunotherapy.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized
- Antibodies, Monoclonal, Murine-Derived
- Antibody-Dependent Cell Cytotoxicity/drug effects
- Antigens, Neoplasm/metabolism
- Antineoplastic Agents, Immunological/pharmacology
- B-Lymphocytes
- Carcinoma, Hepatocellular/metabolism
- Cell Line, Tumor
- Cytophagocytosis/drug effects
- Hep G2 Cells
- Hepatocytes/drug effects
- Hepatocytes/metabolism
- Humans
- Immunotherapy
- Killer Cells, Natural
- Liver Neoplasms/metabolism
- Macrophages
- Mice
- Molecular Targeted Therapy
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/metabolism
- Neoplasm Transplantation
- Neoplasms/metabolism
- Oncogene Proteins/metabolism
- Protein Tyrosine Phosphatases/antagonists & inhibitors
- Protein Tyrosine Phosphatases/metabolism
- Receptors, IgG
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Min Thura
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
| | - Abdul Qader Al-Aidaroos
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
| | - Abhishek Gupta
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
| | - Cheng Ean Chee
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), Singapore, 119082, Singapore
| | - Soo Chin Lee
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), Singapore, 119082, Singapore
| | - Kam Man Hui
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Jie Li
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
| | - Yeoh Khay Guan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119260, Singapore
| | - Wei Peng Yong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), Singapore, 119082, Singapore
| | - Jimmy So
- Division of Surgical Oncology, National University Cancer Institute, Singapore (NCIS), Singapore, 119082, Singapore
| | - Wee Joo Chng
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), Singapore, 119082, Singapore
| | - Chin Hin Ng
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), Singapore, 119082, Singapore
| | - Jianbiao Zhou
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), Singapore, 119082, Singapore
| | - Ling Zhi Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - John Shyi Peng Yuen
- Department of Urology, Singapore General Hospital, Singapore, 169608, Singapore
| | - Henry Sun Sien Ho
- Department of Urology, Singapore General Hospital, Singapore, 169608, Singapore
| | - Sim Mei Yi
- Department of Urology, Singapore General Hospital, Singapore, 169608, Singapore
| | - Edmund Chiong
- Division of Surgical Oncology, National University Cancer Institute, Singapore (NCIS), Singapore, 119082, Singapore
| | - Su Pin Choo
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Joanne Ngeow
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Matthew Chau Hsien Ng
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Clarinda Chua
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Eugene Shen Ann Yeo
- Department of Colorectal Surgery, Singapore General Hospital, Singapore, 169608, Singapore
| | - Iain Bee Huat Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Joel Xuan En Sng
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
| | - Nicholas Yan Zhi Tan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
| | - Jean Paul Thiery
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
| | - Boon Cher Goh
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), Singapore, 119082, Singapore
| | - Qi Zeng
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119260, Singapore.
| |
Collapse
|
56
|
Singh K, Ejaz W, Dutta K, Thayumanavan S. Antibody Delivery for Intracellular Targets: Emergent Therapeutic Potential. Bioconjug Chem 2019; 30:1028-1041. [PMID: 30830750 PMCID: PMC6470022 DOI: 10.1021/acs.bioconjchem.9b00025] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Proteins have sparked fast growing interest as biological therapeutic agents for several diseases. Antibodies, in particular, carry an enormous potential as drugs owing to their remarkable target specificity and low immunogenicity. Although the market has numerous antibodies directed toward extracellular targets, their use in targeting therapeutically important intracellular targets is limited by their inability to cross cellular membrane. Realizing the potential for antibody therapy in disease treatment, progress has been made in the development of methods to deliver antibodies intracellularly. In this review, we address various platforms for delivery of antibodies and their merits and drawbacks.
Collapse
|
57
|
Schlake T, Thess A, Thran M, Jordan I. mRNA as novel technology for passive immunotherapy. Cell Mol Life Sci 2019; 76:301-328. [PMID: 30334070 PMCID: PMC6339677 DOI: 10.1007/s00018-018-2935-4] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 09/13/2018] [Accepted: 10/03/2018] [Indexed: 12/17/2022]
Abstract
While active immunization elicits a lasting immune response by the body, passive immunotherapy transiently equips the body with exogenously generated immunological effectors in the form of either target-specific antibodies or lymphocytes functionalized with target-specific receptors. In either case, administration or expression of recombinant proteins plays a fundamental role. mRNA prepared by in vitro transcription (IVT) is increasingly appreciated as a drug substance for delivery of recombinant proteins. With its biological role as transient carrier of genetic information translated into protein in the cytoplasm, therapeutic application of mRNA combines several advantages. For example, compared to transfected DNA, mRNA harbors inherent safety features. It is not associated with the risk of inducing genomic changes and potential adverse effects are only temporary due to its transient nature. Compared to the administration of recombinant proteins produced in bioreactors, mRNA allows supplying proteins that are difficult to manufacture and offers extended pharmacokinetics for short-lived proteins. Based on great progress in understanding and manipulating mRNA properties, efficacy data in various models have now demonstrated that IVT mRNA constitutes a potent and flexible platform technology. Starting with an introduction into passive immunotherapy, this review summarizes the current status of IVT mRNA technology and its application to such immunological interventions.
Collapse
Affiliation(s)
- Thomas Schlake
- CureVac AG, Paul-Ehrlich-Str. 15, 72076, Tübingen, Germany.
| | - Andreas Thess
- CureVac AG, Paul-Ehrlich-Str. 15, 72076, Tübingen, Germany
| | - Moritz Thran
- CureVac AG, Paul-Ehrlich-Str. 15, 72076, Tübingen, Germany
| | - Ingo Jordan
- CureVac AG, Paul-Ehrlich-Str. 15, 72076, Tübingen, Germany
| |
Collapse
|
58
|
Slastnikova TA, Ulasov AV, Rosenkranz AA, Sobolev AS. Targeted Intracellular Delivery of Antibodies: The State of the Art. Front Pharmacol 2018; 9:1208. [PMID: 30405420 PMCID: PMC6207587 DOI: 10.3389/fphar.2018.01208] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 10/03/2018] [Indexed: 12/11/2022] Open
Abstract
A dominant area of antibody research is the extension of the use of this mighty experimental and therapeutic tool for the specific detection of molecules for diagnostics, visualization, and activity blocking. Despite the ability to raise antibodies against different proteins, numerous applications of antibodies in basic research fields, clinical practice, and biotechnology are restricted to permeabilized cells or extracellular antigens, such as membrane or secreted proteins. With the exception of small groups of autoantibodies, natural antibodies to intracellular targets cannot be used within living cells. This excludes the scope of a major class of intracellular targets, including some infamous cancer-associated molecules. Some of these targets are still not druggable via small molecules because of large flat contact areas and the absence of deep hydrophobic pockets in which small molecules can insert and perturb their activity. Thus, the development of technologies for the targeted intracellular delivery of antibodies, their fragments, or antibody-like molecules is extremely important. Various strategies for intracellular targeting of antibodies via protein-transduction domains or their mimics, liposomes, polymer vesicles, and viral envelopes, are reviewed in this article. The pitfalls, challenges, and perspectives of these technologies are discussed.
Collapse
Affiliation(s)
- Tatiana A. Slastnikova
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - A. V. Ulasov
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - A. A. Rosenkranz
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
- Faculty of Biology, M. V. Lomonosov Moscow State University, Moscow, Russia
| | - A. S. Sobolev
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
- Faculty of Biology, M. V. Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
59
|
Park H, Kim M, Seo Y, Ham Y, Cho MY, Kwon MH. Cytosolic Internalization of Anti-DNA Antibodies by Human Monocytes Induces Production of Pro-inflammatory Cytokines Independently of the Tripartite Motif-Containing 21 (TRIM21)-Mediated Pathway. Front Immunol 2018; 9:2019. [PMID: 30233598 PMCID: PMC6131520 DOI: 10.3389/fimmu.2018.02019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 08/16/2018] [Indexed: 01/05/2023] Open
Abstract
Anti-DNA autoantibodies are a hallmark of systemic lupus erythematosus (SLE). A subset of anti-DNA IgG autoantibodies is cell-internalizable; thus they can enter living cells in the form of free IgG. However, the contribution made by the Fc region of internalized free-form IgG to the cytokine response has not been studied, despite the recent discovery of tripartite motif-containing 21 (TRIM21), a cytosolic Fc receptor involved in immune signaling. This study used an internalizable IgG anti-DNA antibody (3D8) to examine the cytokine responses of human monocytes to the Fc region of cytosolic free IgG. Internalization of 3D8 IgG and a 3D8 single-chain variable fragment-Fc (scFv-Fc) induced production of IL-8 and TNF-α via activation of NF-κB. By contrast, a 3D8 scFv (comprising variable domains alone) did not. This suggests Fc-dependent cytokine signaling. A 3D8 IgG-N434D mutant that is not recognized by TRIM21 induced greater production of cytokines than 3D8 IgG. Moreover the amounts of cytokines induced by 3D8 IgG in TRIM21-knockdown THP-1 cells were higher than those in control cells, indicating that cytokine signaling is not mediated by TRIM21. The results suggest the existence of a novel Fc-dependent signaling pathway that is activated upon internalization of IgG antibodies by human monocytes.
Collapse
Affiliation(s)
- Hyunjoon Park
- Department of Biomedical Sciences, Graduate School, Ajou University, Yeongtong-gu, Suwon, South Korea.,Department of Microbiology, Ajou University School of Medicine, Suwon, South Korea
| | - Minjae Kim
- Department of Biomedical Sciences, Graduate School, Ajou University, Yeongtong-gu, Suwon, South Korea.,Department of Microbiology, Ajou University School of Medicine, Suwon, South Korea
| | - Youngsil Seo
- Department of Biomedical Sciences, Graduate School, Ajou University, Yeongtong-gu, Suwon, South Korea.,Department of Microbiology, Ajou University School of Medicine, Suwon, South Korea
| | - Yeonkyoung Ham
- Department of Biomedical Sciences, Graduate School, Ajou University, Yeongtong-gu, Suwon, South Korea.,Department of Microbiology, Ajou University School of Medicine, Suwon, South Korea
| | - Mi-Young Cho
- Department of Biomedical Sciences, Graduate School, Ajou University, Yeongtong-gu, Suwon, South Korea.,Department of Microbiology, Ajou University School of Medicine, Suwon, South Korea
| | - Myung-Hee Kwon
- Department of Biomedical Sciences, Graduate School, Ajou University, Yeongtong-gu, Suwon, South Korea.,Department of Microbiology, Ajou University School of Medicine, Suwon, South Korea
| |
Collapse
|