1
|
Li W, Sun J, Feng S, La Rosa A, Zhang P, Wu EY, Loeser R, Li C. Secreted PD-L1 alleviates inflammatory arthritis in mice through local and systemic AAV gene therapy. Front Immunol 2025; 16:1527858. [PMID: 39963137 PMCID: PMC11830590 DOI: 10.3389/fimmu.2025.1527858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Introduction Rheumatoid arthritis (RA) primarily affects the joints but can also affect multiple organs and profoundly impacts patients' ability to carry out daily activities, mental health, and life expectancy. Current treatments for RA are limited in terms of duration, efficacy, and adverse effects. PD-L1 is a checkpoint protein that plays important roles in immune regulation and has been implicated in the initiation and progression of multiple autoimmune diseases. Method In a previous study, we demonstrated that intra-articular injection with adeno-associated virus (AAV) vectors encoding wild type PD-L1 improved local inflammation in the joint in the collagen-induced arthritis (CIA) mouse model of RA. To further improve efficacy, we explored AAV-mediated delivery of the soluble PD-L1 (sPD-L1) to CIA mice. Result After intra-articular injection of AAV6 vectors expressing the optimal isoform of sPD-L1 (shPD-L1), more potency was observed when compared to wild type PD-L1, with a lower dose of AAV6/shPD-L1 needed for arthritis improvement. To study the therapeutic effect of systemic expression of sPD-L1, we administered AAV8/shPD-L1 gene therapy in CIA mice via retro-orbital injection and found significant improvements in joint inflammation and paw swelling, exhibiting similar phenotypes to that in naïve mice. The levels of total immunoglobulin and anti-collagen specific antibodies were lower in AAV8/shPD-L1 treated CIA mice than those in controls. The levels of pro-inflammatory cytokines in blood were also significantly decreased in shPD-L1 treated mice. Additionally, T cell apoptosis rates in the spleen showed a 2-fold increase in treated mice. Finally, we investigated the therapeutic effect of AAV/shPD-L1 via intramuscular injection. After injection of AAV6/shPD-L1, decreased paw swelling, reduced joint inflammation, and lower levels of pro-inflammatory cytokines in blood were achieved. The therapeutic effect of shPD-L1 was dose dependent via intramuscular treatment with AAV vectors. Conclusion In conclusion, the findings in this study suggest that intra-articular injection of AAV vectors encoding sPD-L1 results in greater therapeutic benefit on arthritis, and systemic AAV/sPD-L1 is able to block the development of inflammatory arthritis with inhibition of the systemic immune response, underlining the potential of gene therapy with systemic delivery of shPD-L1 via AAV vectors in RA.
Collapse
Affiliation(s)
- Wenjun Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Division of Oral and Craniofacial Biomedicine, University of North Carolina Adams School of Dentistry, Chapel Hill, NC, United States
| | - Junjiang Sun
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Susi Feng
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ariana La Rosa
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Panli Zhang
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Eveline Y. Wu
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Richard Loeser
- Division of Rheumatology, Allergy, and Immunology, University of North Carolina, Chapel Hill, NC, United States
| | - Chengwen Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
2
|
Rotondo C, Perniola S, Parisi S, Cantatore FP, Corrado A. Target Therapy in Chronic Arthritis: The Unmet Needs, State-of-the-Art on Dual Biologic Treatments, and Future Perspectives. J Clin Med 2024; 13:7303. [PMID: 39685761 DOI: 10.3390/jcm13237303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 11/12/2024] [Indexed: 12/18/2024] Open
Abstract
Since the early 1990s, the introduction of biologic disease-modifying antirheumatic drugs (b-DMARDs) in managing rheumatological diseases has revolutionised the course of inflammatory chronic arthritis, improving the quality of life, slowing the radiographic progression, avoiding disability, preserving workability, and reducing mortality [...].
Collapse
Affiliation(s)
- Cinzia Rotondo
- Rheumatology Unit, Department of Medical and Surgical Sciences, Azienda Ospedaliero-Universitaria Policlinico Riuniti di Foggia, Università degli Studi di Foggia, 71121 Foggia, Italy
| | - Simone Perniola
- Rheumatology Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari "Aldo Moro", 70125 Bari, Italy
| | - Simone Parisi
- Rheumatology Unit, Azienda Ospedaliera Universitaria Città della Salute e delle Scienza di Torino, 10126 Torino, Italy
| | - Francesco Paolo Cantatore
- Rheumatology Unit, Department of Medical and Surgical Sciences, Azienda Ospedaliero-Universitaria Policlinico Riuniti di Foggia, Università degli Studi di Foggia, 71121 Foggia, Italy
| | - Addolorata Corrado
- Rheumatology Unit, Department of Medical and Surgical Sciences, Azienda Ospedaliero-Universitaria Policlinico Riuniti di Foggia, Università degli Studi di Foggia, 71121 Foggia, Italy
| |
Collapse
|
3
|
Rose-John S, Jenkins BJ, Garbers C, Moll JM, Scheller J. Targeting IL-6 trans-signalling: past, present and future prospects. Nat Rev Immunol 2023; 23:666-681. [PMID: 37069261 PMCID: PMC10108826 DOI: 10.1038/s41577-023-00856-y] [Citation(s) in RCA: 187] [Impact Index Per Article: 93.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2023] [Indexed: 04/19/2023]
Abstract
Interleukin-6 (IL-6) is a key immunomodulatory cytokine that affects the pathogenesis of diverse diseases, including autoimmune diseases, chronic inflammatory conditions and cancer. Classical IL-6 signalling involves the binding of IL-6 to the membrane-bound IL-6 receptor α-subunit (hereafter termed 'mIL-6R') and glycoprotein 130 (gp130) signal-transducing subunit. By contrast, in IL-6 trans-signalling, complexes of IL-6 and the soluble form of IL-6 receptor (sIL-6R) signal via membrane-bound gp130. A third mode of IL-6 signalling - known as cluster signalling - involves preformed complexes of membrane-bound IL-6-mIL-6R on one cell activating gp130 subunits on target cells. Antibodies and small molecules have been developed that block all three forms of IL-6 signalling, but in the past decade, IL-6 trans-signalling has emerged as the predominant pathway by which IL-6 promotes disease pathogenesis. The first selective inhibitor of IL-6 trans-signalling, sgp130, has shown therapeutic potential in various preclinical models of disease and olamkicept, a sgp130Fc variant, had promising results in phase II clinical studies for inflammatory bowel disease. Technological developments have already led to next-generation sgp130 variants with increased affinity and selectivity towards IL-6 trans-signalling, along with indirect strategies to block IL-6 trans-signalling. Here, we summarize our current understanding of the biological outcomes of IL-6-mediated signalling and the potential for targeting this pathway in the clinic.
Collapse
Affiliation(s)
- Stefan Rose-John
- Biochemical Institute, Medical Faculty, Christian-Albrechts-University, Kiel, Germany
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Christoph Garbers
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GC:I3), Otto-von-Guericke-University, Magdeburg, Germany
- Center for Health and Medical Prevention (CHaMP), Otto-von-Guericke-University, Magdeburg, Germany
| | - Jens M Moll
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany.
| |
Collapse
|
4
|
Biesemann N, Margerie D, Asbrand C, Rehberg M, Savova V, Agueusop I, Klemmer D, Ding-Pfennigdorff D, Schwahn U, Dudek M, Heyninck K, De Tavernier E, Cornelis S, Kohlmann M, Nestle FO, Herrmann M. Additive efficacy of a bispecific anti-TNF/IL-6 nanobody compound in translational models of rheumatoid arthritis. Sci Transl Med 2023; 15:eabq4419. [PMID: 36724239 DOI: 10.1126/scitranslmed.abq4419] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Rheumatoid arthritis (RA) is one of the most common autoimmune diseases affecting primarily the joints. Despite successful therapies including antibodies against tumor necrosis factor (TNF) and interleukin-6 (IL-6) receptor, only 20 to 30% of patients experience remission. We studied whether inhibiting both TNF and IL-6 would result in improved efficacy. Using backtranslation from single-cell RNA sequencing (scRNA-seq) data from individuals with RA, we hypothesized that TNF and IL-6 act synergistically on fibroblast-like synoviocytes (FLS) and T cells. Coculture of FLS from individuals with RA and T cells supported this hypothesis, revealing effects on both disease-driving pathways and biomarkers. Combining anti-TNF and anti-IL-6 antibodies in collagen-induced arthritis (CIA) mouse models resulted in sustained long-term remission, improved histology, and effects on bone remodeling pathways. These promising data initiated the development of an anti-TNF/IL-6 bispecific nanobody compound 1, with similar potencies against TNF and IL-6. We observed additive efficacy of compound 1 in a FLS/T cell coculture affecting arthritis and T helper 17 (TH17) pathways. This nanobody compound transcript signature inversely overlapped with described RA endotypes, indicating a potential efficacy in a broader patient population. In summary, we showed superiority of a bispecific anti-TNF/IL-6 nanobody compound or combination treatment over monospecific treatments in both in vitro and in vivo models. We anticipate improved efficacy in upcoming clinical studies.
Collapse
Affiliation(s)
- Nadine Biesemann
- Sanofi R&D, Immunology and Inflammation Therapeutic Area, Type 1/17 Immunology Cluster, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Daniel Margerie
- Sanofi R&D, Digital and Data Science, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Christian Asbrand
- Sanofi R&D, Immunology and Inflammation Therapeutic Area, Type 1/17 Immunology Cluster, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Markus Rehberg
- Sanofi R&D, Digital and Data Science, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Virginia Savova
- Precision Medicine and Computational Biology, Sanofi R&D, 350 Water St., Cambridge, MA 02141, USA
| | - Inoncent Agueusop
- Sanofi R&D, Biostatistics and Programming, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Daniel Klemmer
- Sanofi R&D, Biostatistics and Programming, Non-Clinical Efficacy and Safety, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Danping Ding-Pfennigdorff
- Sanofi R&D, Immunology and Inflammation Therapeutic Area, Type 1/17 Immunology Cluster, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Uwe Schwahn
- Sanofi R&D, Translational Medicine and Early Development, Biomarkers and Clinical Bioanalysis, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Markus Dudek
- Sanofi R&D, Translational Medicine and Early Development, Biomarkers and Clinical Bioanalysis, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Karen Heyninck
- Sanofi R&D, NANOBODY Research Platform, Technologiepark 21, 9052 Zwijnaarde, Belgium
| | - Evelyn De Tavernier
- Sanofi R&D, NANOBODY Research Platform, Technologiepark 21, 9052 Zwijnaarde, Belgium
| | - Sigrid Cornelis
- Sanofi R&D, NANOBODY Research Platform, Technologiepark 21, 9052 Zwijnaarde, Belgium
| | - Markus Kohlmann
- Sanofi R&D, Early Clinical Development Therapeutic Area Immunology and Inflammation, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | | | - Matthias Herrmann
- Sanofi R&D, Immunology and Inflammation Therapeutic Area, Type 1/17 Immunology Cluster, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| |
Collapse
|
5
|
Li J, Li YR, Glessner JT, Yang J, March ME, Kao C, Vaccaro CN, Bradfield JP, Li J, Mentch FD, Qu H, Qi X, Chang X, Hou C, Abrams DJ, Qiu H, Wei Z, Connolly JJ, Wang F, Snyder J, Flatø B, Thompson SD, Langefeld CD, Lie BA, Munro JE, Wise C, Sleiman PMA, Hakonarson H. Identification of Novel Loci Shared by Juvenile Idiopathic Arthritis Subtypes Through Integrative Genetic Analysis. Arthritis Rheumatol 2022; 74:1420-1429. [PMID: 35347896 PMCID: PMC9542075 DOI: 10.1002/art.42129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 03/11/2022] [Accepted: 03/25/2022] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Juvenile idiopathic arthritis (JIA) is the most common chronic immune-mediated joint disease among children and encompasses a heterogeneous group of immune-mediated joint disorders classified into 7 subtypes according to clinical presentation. However, phenotype overlap and biologic evidence suggest a shared mechanistic basis between subtypes. This study was undertaken to systematically investigate shared genetic underpinnings of JIA subtypes. METHODS We performed a heterogeneity-sensitive genome-wide association study encompassing a total of 1,245 JIA cases (classified into 7 subtypes) and 9,250 controls, followed by fine-mapping of candidate causal variants at each genome-wide significant locus, functional annotation, and pathway and network analysis. We further identified candidate drug targets and drug repurposing opportunities by in silico analyses. RESULTS In addition to the major histocompatibility complex locus, we identified 15 genome-wide significant loci shared between at least 2 JIA subtypes, including 10 novel loci. Functional annotation indicated that candidate genes at these loci were expressed in diverse immune cell types. CONCLUSION This study identified novel genetic loci shared by JIA subtypes. Our findings identified candidate mechanisms underlying JIA subtypes and candidate targets with drug repurposing opportunities for JIA treatment.
Collapse
Affiliation(s)
- Jin Li
- Department of Cell Biology, the Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical EpigeneticsSchool of Basic Medical Sciences, Tianjin Medical UniversityTianjinChina
| | - Yun R. Li
- The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, City of Hope Comprehensive Cancer Center, Duarte, California, and Translational Genomics Research InstitutePhoenixArizona
| | | | - Jie Yang
- Department of Cell Biology, the Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical EpigeneticsSchool of Basic Medical Sciences, Tianjin Medical UniversityTianjinChina
| | | | - Charlly Kao
- The Children's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | | | | | - Junyi Li
- Department of Cell Biology, the Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical EpigeneticsSchool of Basic Medical Sciences, Tianjin Medical UniversityTianjinChina
| | - Frank D. Mentch
- The Children's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - Hui‐Qi Qu
- The Children's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - Xiaohui Qi
- Department of Cell Biology, the Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical EpigeneticsSchool of Basic Medical Sciences, Tianjin Medical UniversityTianjinChina
| | - Xiao Chang
- The Children's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - Cuiping Hou
- The Children's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - Debra J. Abrams
- The Children's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - Haijun Qiu
- The Children's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - Zhi Wei
- New Jersey Institute of TechnologyNewark
| | | | - Fengxiang Wang
- The Children's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - James Snyder
- The Children's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - Berit Flatø
- Oslo University HospitalRikshospitaletOsloNorway
| | - Susan D. Thompson
- University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical CenterCincinnatiOhio
| | - Carl D. Langefeld
- Wake Forest University School of MedicineWinston‐SalemNorth Carolina
| | | | | | | | | | - Hakon Hakonarson
- The Children's Hospital of Philadelphia and University of PennsylvaniaPhiladelphia
| |
Collapse
|
6
|
Muench DE, Sun Z, Sharma A, Tang C, Crampton JS, Lao C, Kersjes K, Chang W, Na S. A Pathogenic Th17/CD38 + Macrophage Feedback Loop Drives Inflammatory Arthritis through TNF-α. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1315-1328. [PMID: 35197330 DOI: 10.4049/jimmunol.2101025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/04/2022] [Indexed: 12/29/2022]
Abstract
The pathobiology of rheumatoid inflammatory diseases, including rheumatoid arthritis (RA) and psoriatic arthritis, involves the interplay between innate and adaptive immune components and resident synoviocytes. Single-cell analyses of patient samples and relevant mouse models have characterized many cellular subsets in RA. However, the impact of interactions between cell types is not fully understood. In this study, we temporally profiled murine arthritic synovial isolates at the single-cell level to identify perturbations similar to those found in human RA. Notably, murine macrophage subtypes like those found in RA patients were expanded in arthritis and linked to promoting the function of Th17 cells in the joint. In vitro experiments identified a capacity for murine macrophages to maintain the functionality and expansion of Th17 cells. Reciprocally, murine Th17 cell-derived TNF-α induced CD38+ macrophages that enhanced Th17 functionality. Murine synovial CD38+ macrophages were expanded during arthritis, and their depletion or blockade via TNF-α neutralization alleviated disease while reducing IL-17A-producing cells. These findings identify a cellular feedback loop that promotes Th17 cell pathogenicity through TNF-α to drive inflammatory arthritis.
Collapse
Affiliation(s)
- David E Muench
- Immunology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, San Diego, CA
| | - Zhe Sun
- Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN; and
| | - Anchal Sharma
- Research Information and Digital Solutions, Lilly Research Laboratories, Eli Lilly and Company, New York, NY
| | - Crystal Tang
- Immunology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, San Diego, CA
| | - Jordan S Crampton
- Immunology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, San Diego, CA
| | - Christopher Lao
- Immunology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, San Diego, CA
| | - Kara Kersjes
- Immunology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, San Diego, CA
| | - William Chang
- Immunology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, San Diego, CA
| | - Songqing Na
- Immunology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, San Diego, CA;
| |
Collapse
|
7
|
Minicircles for Investigating and Treating Arthritic Diseases. Pharmaceutics 2021; 13:pharmaceutics13050736. [PMID: 34067675 PMCID: PMC8156692 DOI: 10.3390/pharmaceutics13050736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/10/2021] [Accepted: 05/15/2021] [Indexed: 01/22/2023] Open
Abstract
Gene delivery systems have become an essential component of research and the development of therapeutics for various diseases. Minicircles are non-viral vectors with promising characteristics for application in a variety of fields. With their minimal size, minicircles exhibit relatively high safety and efficient delivery of genes of interest into cells. Cartilage tissue lacks the natural ability to heal, making it difficult to treat osteoarthritis (OA) and rheumatoid arthritis (RA), which are the two main types of joint-related disease. Although both OA and RA affect the joint, RA is an autoimmune disease, while OA is a degenerative joint condition. Gene transfer using minicircles has also been used in many studies regarding cartilage and its diseased conditions. In this review, we summarize the cartilage-, OA-, and RA-based studies that have used minicircles as the gene delivery system.
Collapse
|
8
|
Ando T, Yamamoto M, Takamori Y, Tsukamoto K, Fuji D, Kawakami T. In vitro selection of an RNA aptamer yields an interleukin-6/interleukin-6 receptor interaction inhibitor. Biosci Biotechnol Biochem 2021; 85:1170-1174. [PMID: 33590853 PMCID: PMC7799299 DOI: 10.1093/bbb/zbaa124] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 12/29/2020] [Indexed: 12/13/2022]
Abstract
Interleukin-6 (IL-6) binds to IL-6 receptor (IL-6R) subunit, related to autoimmune diseases and cytokine storm in COVID-19. In this study we performed Systematic Evolution of Ligands by Exponential enrichment (SELEX) and identified a novel RNA aptamer. This RNA aptamer not only bound to IL-6R with a dissociation constant of 200 nM, but also inhibited the interaction of IL-6R with IL-6.
Collapse
Affiliation(s)
- Takehiro Ando
- Department of Life and Environmental Sciences, Integrated Graduate School of Medicine, Engineering, and Agricultural Sciences, University of Yamanashi, Kofu, Yamanashi, Japan
| | - Mizuki Yamamoto
- Department of Integrated Applied Life Science, Integrated Graduate School of Medicine, Engineering, and Agricultural Sciences, University of Yamanashi, Kofu, Yamanashi, Japan
| | - Yukio Takamori
- Department of Life and Environmental Sciences, Integrated Graduate School of Medicine, Engineering, and Agricultural Sciences, University of Yamanashi, Kofu, Yamanashi, Japan
| | - Keita Tsukamoto
- Department of Life and Environmental Sciences, Integrated Graduate School of Medicine, Engineering, and Agricultural Sciences, University of Yamanashi, Kofu, Yamanashi, Japan
| | - Daisuke Fuji
- Department of Biotechnology, Faculty of Life and Environmental Sciences, University of Yamanashi, Kofu, Yamanashi, Japan
| | - Takashi Kawakami
- Faculty of Life and Environmental Sciences, Graduate Faculty of Interdisciplinary Research, University of Yamanashi, Kofu, Yamanashi, Japan.,JST, PRESTO, Kawaguchi, Saitama, Japan
| |
Collapse
|
9
|
Chen X, Bian Y, Xie Y, Zheng N, Nie K, Liu R, Yan M, Luo H, Wang H, Yang J, Zhang N. A Dual Target-Directed Single Domain-Based Fusion Protein Against Interleukin-6 Receptor Decelerate Experimental Arthritis Progression Via Modulating JNK Expression. Inflammation 2021; 44:1620-1628. [PMID: 33751358 PMCID: PMC7943332 DOI: 10.1007/s10753-021-01446-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 02/07/2021] [Accepted: 03/01/2021] [Indexed: 12/11/2022]
Abstract
The currently used anti-cytokine therapeutic antibodies cannot selectively neutralize pathogenic cytokine signalling that cause collateral damage to protective signalling cascades. The single domain chain firstly discovered in Camelidae displays fully functional ability in antigen-binding against variable targets, which has been seemed as attractive candidates for the next-generation biologic drug study. In this study, we established a simple prokaryotic expression system for a dual target-directed single domain-based fusion protein against the interleukin-6 receptor and human serum, albumin, the recombinant anti-IL-6R fusion protein (VHH-0031). VHH-0031 exhibited potent anti-inflammatory effects produced by LPS on cell RAW264.7, where the major cytokines and NO production were downregulated after 24 h incubation with VHH-0031 in a dose-dependent manner. In vivo, VHH-0031 presented significant effects on the degree reduction of joint swelling in the adjuvant-induced arthritis (AIA) rat, having a healthier appearance compared with the dexamethasone. The expression level of JNK protein in the VHH-0031 group was significantly decreased, demonstrating that VHH-0031 provides a low-cost and desirable effect in the treatment of more widely patients.
Collapse
Affiliation(s)
- Xiaole Chen
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian, China
| | - Yize Bian
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Yongqing Xie
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Ningning Zheng
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Kaimei Nie
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Rui Liu
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Mengru Yan
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Hongbin Luo
- Department of Orthopedics,The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
| | - He Wang
- School of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China.
| | - Juhua Yang
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China. .,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian, China.
| | - Nanwen Zhang
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian, China. .,Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
10
|
Chen X, Nie K, Zhang X, Tan S, Zheng Q, Wang Y, Chen X, Tang Z, Liu R, Yan M, Liu Z, Lin J, Xu J, Zhang N, Wang H, Yang J. The recombinant anti-TNF-α fusion protein ameliorates rheumatoid arthritis by the protective role of autophagy. Biosci Rep 2020; 40:BSR20194515. [PMID: 32880389 PMCID: PMC7502693 DOI: 10.1042/bsr20194515] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 08/31/2020] [Accepted: 09/02/2020] [Indexed: 01/08/2023] Open
Abstract
The currently used anti-cytokine therapeutic antibodies cannot selectively neutralize pathogenic cytokine signaling that cause collateral damage to protective signaling cascades carrying the potential for unwanted side effects. The variable domains of heavy-chain only antibodies (HCAbs) discovered in Camelidae are stable and display to be fully functional in antigen-binding against variable targets, which seem to be attractive candidates for the next-generation biologic drug study. The purpose of our study was to establish a simple prokaryotic expression system for large-scale expression, purification, and refolding of the recombinant anti-tumor necrosis factor α (TNF-α) fusion protein (FVH1-1) from inclusion bodies. Over 95% purity of the recombinant anti-TNF-α fusion proteins was obtained by just one purification step in our developed prokaryotic expression system, while the results of surface plasmon resonance (SPR) established the high-efficiency potent binding ability of FVH1-1 to human TNF-α. The counteraction of TNF-α cytotoxic effect experiment on the mouse fibroblast fibrosarcoma cell line (L929) confirmed that the expressed FVH1-1 were able to selectively and highly combine with human recombinant TNF-α (hTNF-α) in vitro. Western blot results showed that FVH1-1 can inhibit the activation of caspase-9 and PARP, which are the apoptotic signaling pathway proteins activated by hTNF-α. Meanwhile, lysosome autophagy signaling pathways stimulated by hTNF-α were inhibited by FVH1-1, which down-regulated the expression of LC3II/LC3I and up-regulated the expression of P62, indicating that the autophagy linked with TNF-α-induced apoptosis in response to rheumatoid arthritis. The results of the AIA rat model experiment presented that FVH1-1 can reduce the degree of joint swelling and inflammatory factors to a certain extent in vivo.
Collapse
Affiliation(s)
- Xiaole Chen
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian, China
| | - Kaimei Nie
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Xin Zhang
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Shuangyu Tan
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Qingmei Zheng
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Yaduan Wang
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Xiaofeng Chen
- Department of Neurorehabilitation, Rehabilitation Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Zhiyu Tang
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Rui Liu
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Mengru Yan
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhiwei Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Jianbo Lin
- Department of Thoracic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Jianhua Xu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Nanwen Zhang
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian, China
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - He Wang
- School of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Juhua Yang
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
11
|
Drutskaya MS, Nosenko MA, Gorshkova EA, Mokhonov VV, Zvartsev RV, Polinova AI, Kruglov AA, Nedospasov SA. Effects of myeloid cell-restricted TNF inhibitors in vitro and in vivo. J Leukoc Biol 2020; 107:933-939. [PMID: 32040234 DOI: 10.1002/jlb.3ab0120-532r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/01/2020] [Accepted: 01/18/2020] [Indexed: 12/17/2022] Open
Abstract
Systemic TNF neutralization can be used as a therapy for several autoimmune diseases. To evaluate the effects of cell type-restricted TNF blockade, we previously generated bispecific antibodies that can limit TNF secretion by myeloid cells (myeloid cell-specific TNF inhibitors or MYSTIs). In this study several such variable domain (VH) of a camelid heavy-chain only antibody-based TNF inhibitors were compared in relevant experimental models, both in vitro and in vivo. Pretreatment with MYSTI-2, containing the anti-F4/80 module, can restrict the release of human TNF (hTNF) from LPS-activated bone marrow-derived macrophage (BMDM) cultures of humanized TNF knock-in (mice; hTNFKI) more effectively than MYSTI-3, containing the anti-CD11b module. MYSTI-2 was also superior to MYSTI-3 in providing in vivo protection in acute toxicity model. Finally, MYSTI-2 was at least as effective as Infliximab in preventing collagen antibody-induced arthritis. This study demonstrates that a 33 kDa bispecific mini-antibody that specifically restricts TNF secretion by macrophages is efficient for amelioration of experimental arthritis.
Collapse
Affiliation(s)
- Marina S Drutskaya
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Maxim A Nosenko
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Ekaterina A Gorshkova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,Lomonosov Moscow State University, Moscow, Russia
| | - Vladislav V Mokhonov
- Blokhina Scientific Research Institute of Epidemiology and Microbiology of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Ruslan V Zvartsev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | | | - Andrey A Kruglov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,Lomonosov Moscow State University, Moscow, Russia.,German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Sergei A Nedospasov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,Lomonosov Moscow State University, Moscow, Russia.,Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
12
|
Bispecific Antibodies for Autoimmune and Inflammatory Diseases: Clinical Progress to Date. BioDrugs 2020; 34:111-119. [DOI: 10.1007/s40259-019-00400-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
13
|
Park J, Yoo S, Lim M, Ryu J, Oh H, Hwang S, Yang S, Jung K, Yoon S, Park B, Park S, Kim H, Cho M, Park Y. A bispecific soluble receptor fusion protein that targets TNF‐α and IL‐21 for synergistic therapy in inflammatory arthritis. FASEB J 2019; 34:248-262. [DOI: 10.1096/fj.201900816rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 09/29/2019] [Accepted: 10/08/2019] [Indexed: 01/20/2023]
Affiliation(s)
- Jin‐Sil Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine The Catholic University of Korea Seoul Republic of Korea
| | | | - Mi‐Ae Lim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine The Catholic University of Korea Seoul Republic of Korea
| | - Jun‐Geol Ryu
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine The Catholic University of Korea Seoul Republic of Korea
| | - Hye‐Joa Oh
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine The Catholic University of Korea Seoul Republic of Korea
| | - Sun‐Hee Hwang
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine The Catholic University of Korea Seoul Republic of Korea
| | - SeungCheon Yang
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine The Catholic University of Korea Seoul Republic of Korea
| | - Kyung‐Ah Jung
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine The Catholic University of Korea Seoul Republic of Korea
| | | | | | - Sung‐Hwan Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine The Catholic University of Korea Seoul Republic of Korea
- Divison of Rheumatology, Department of Internal Medicine Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea Seoul Republic of Korea
| | - Ho‐Youn Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine The Catholic University of Korea Seoul Republic of Korea
| | - Mi‐La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine The Catholic University of Korea Seoul Republic of Korea
- Department of Biomedicine & Health Sciences, College of Medicine The Catholic University of Korea Seoul Republic of Korea
- Department of Medical Lifescience, College of Medicine The Catholic University of Korea Seoul Republic of Korea
| | | |
Collapse
|
14
|
Boleto G, Kanagaratnam L, Dramé M, Salmon JH. Safety of combination therapy with two bDMARDs in patients with rheumatoid arthritis: A systematic review and meta-analysis. Semin Arthritis Rheum 2018; 49:35-42. [PMID: 30638975 DOI: 10.1016/j.semarthrit.2018.12.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 11/25/2018] [Accepted: 12/12/2018] [Indexed: 12/28/2022]
Abstract
OBJECTIVES We performed a systematic review and meta-analysis of the current literature to assess the safety of combining two biologic disease-modifying antirheumatic drugs (bDMARDs) in the treatment of rheumatoid arthritis (RA). METHODS We systematically searched for controlled studies evaluating safety in patients with RA treated with two bDMARDs independently of dose-regimen. Databases used were MEDLINE (via Pubmed), EMBase, Cochrane Library, Scopus, ClinicalTrials.gov, and the WHO International Clinical Trials Registry platform. A meta-analysis was performed between groups on combination therapy and patients on single therapy using random effects model calculating odds ratio (OR) as well as 95% confidence interval (CI). The primary outcome was the rate of serious adverse events (SAEs). RESULTS Six studies with a total of 623 patients (410 on combination therapy and 213 on single therapy) were included. Median follow-up was 9.5 months (range 6-12 months). There was a significant increase in SAEs in the combination group (14.9 vs 6.0%, OR 2.51, 95% CI 1.29-4.89, I2 0%) as well as in total adverse events (94.6 vs 89.1%, OR 2.07, 95% CI 1.11-3.86, I2 0%). When performing subgroup analysis in patients receiving only full-dose of both bDMARDs there was a significant increase in serious infections (6.7 vs 0.6%, OR 5.58, 95% CI 1.25-24.90, I2 0%) and the risk of SAEs remained significantly higher (17.1 vs 6.2%, OR 2.72, 95% CI 1.30-5.69, I2 0%). CONCLUSION Our findings suggest that combination therapy with two bDMARDs in RA appears to increase the risk of SAEs during the first twelve months of treatment.
Collapse
Affiliation(s)
- Gonçalo Boleto
- Rheumatology Department, Maison Blanche Hospital, Reims University Hospitals, 45 Rue Cognacq-Jay, Reims 51092, France
| | - Lukshe Kanagaratnam
- Department of Research and Innovation, Robert Debré Hospital, Reims University Hospitals, 51092 Reims, France; Faculty of Medicine, EA 3797, University of Reims Champagne-Ardenne, 51095 Reims, France
| | - Moustapha Dramé
- Department of Research and Innovation, Robert Debré Hospital, Reims University Hospitals, 51092 Reims, France; Faculty of Medicine, EA 3797, University of Reims Champagne-Ardenne, 51095 Reims, France
| | - Jean-Hugues Salmon
- Rheumatology Department, Maison Blanche Hospital, Reims University Hospitals, 45 Rue Cognacq-Jay, Reims 51092, France; Faculty of Medicine, EA 3797, University of Reims Champagne-Ardenne, 51095 Reims, France.
| |
Collapse
|
15
|
Abstract
Bispecific antibodies have moved from being an academic curiosity with therapeutic promise to reality, with two molecules being currently commercialized (Hemlibra® and Blincyto®) and many more in clinical trials. The success of bispecific antibodies is mainly due to the continuously growing number of mechanisms of actions (MOA) they enable that are not accessible to monoclonal antibodies. One of the earliest MOA of bispecific antibodies and currently the one with the largest number of clinical trials is the redirecting of the cytotoxic activity of T-cells for oncology applications, now extending its use in infective diseases. The use of bispecific antibodies for crossing the blood-brain barrier is another important application because of its potential to advance the therapeutic options for neurological diseases. Another noteworthy application due to its growing trend is enabling a more tissue-specific delivery or activity of antibodies. The different molecular solutions to the initial hurdles that limited the development of bispecific antibodies have led to the current diverse set of bispecific or multispecific antibody formats that can be grouped into three main categories: IgG-like formats, antibody fragment-based formats, or appended IgG formats. The expanded applications of bispecific antibodies come at the price of additional challenges for clinical development. The rising complexity in their structure may increase the risk of immunogenicity and the multiple antigen specificity complicates the selection of relevant species for safety assessment.
Collapse
Affiliation(s)
- Bushra Husain
- Protein Chemistry Department, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Diego Ellerman
- Protein Chemistry Department, Genentech Inc., South San Francisco, CA, 94080, USA.
| |
Collapse
|
16
|
A novel fusion protein attenuates collagen–induced arthritis by targeting interleukin 17A and tumor necrosis factor α. Int J Pharm 2018; 547:72-82. [DOI: 10.1016/j.ijpharm.2018.05.058] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 05/19/2018] [Accepted: 05/23/2018] [Indexed: 12/27/2022]
|
17
|
A New Venue of TNF Targeting. Int J Mol Sci 2018; 19:ijms19051442. [PMID: 29751683 PMCID: PMC5983675 DOI: 10.3390/ijms19051442] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 04/25/2018] [Accepted: 05/03/2018] [Indexed: 12/20/2022] Open
Abstract
The first Food and Drug Administration-(FDA)-approved drugs were small, chemically-manufactured and highly active molecules with possible off-target effects, followed by protein-based medicines such as antibodies. Conventional antibodies bind a specific protein and are becoming increasingly important in the therapeutic landscape. A very prominent class of biologicals are the anti-tumor necrosis factor (TNF) drugs that are applied in several inflammatory diseases that are characterized by dysregulated TNF levels. Marketing of TNF inhibitors revolutionized the treatment of diseases such as Crohn’s disease. However, these inhibitors also have undesired effects, some of them directly associated with the inherent nature of this drug class, whereas others are linked with their mechanism of action, being pan-TNF inhibition. The effects of TNF can diverge at the level of TNF format or receptor, and we discuss the consequences of this in sepsis, autoimmunity and neurodegeneration. Recently, researchers tried to design drugs with reduced side effects. These include molecules with more specificity targeting one specific TNF format or receptor, or that neutralize TNF in specific cells. Alternatively, TNF-directed biologicals without the typical antibody structure are manufactured. Here, we review the complications related to the use of conventional TNF inhibitors, together with the anti-TNF alternatives and the benefits of selective approaches in different diseases.
Collapse
|
18
|
β2-adrenoceptor signaling reduction is involved in the inflammatory response of fibroblast-like synoviocytes from adjuvant-induced arthritic rats. Inflammopharmacology 2018; 27:271-279. [DOI: 10.1007/s10787-018-0477-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 03/28/2018] [Indexed: 12/21/2022]
|
19
|
Jeong SH, Nam Y, Jung H, Kim J, Rim YA, Park N, Lee K, Choi S, Jang Y, Kim Y, Moon JH, Jung SM, Park SH, Ju JH. Interrupting oral infection of Porphyromonas gingivalis with anti-FimA antibody attenuates bacterial dissemination to the arthritic joint and improves experimental arthritis. Exp Mol Med 2018; 50:e460. [PMID: 29568073 PMCID: PMC5898898 DOI: 10.1038/emm.2017.301] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 08/29/2017] [Accepted: 09/26/2017] [Indexed: 12/29/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that typically results in strong inflammation and bone destruction in the joints. It is generally known that the pathogenesis of RA is linked to cardiovascular and periodontal diseases. Though rheumatoid arthritis and periodontitis share many pathologic features such as a perpetual inflammation and bone destruction, the precise mechanism underlying a link between these two diseases has not been fully elucidated. Collagen-induced arthritis (CIA) mice were orally infected with Porphyromonas gingivalis (Pg) or Pg preincubated with an anti-FimA antibody (FimA Ab) specific for fimbriae that are flexible appendages on the cell surface. Pg-infected CIA mice showed oral microbiota disruption and increased alveolar bone loss and had synovitis and joint bone destruction. However, preincubation with FimA Ab led to a significant reduction in the severity of both oral disease and arthritis. Moreover, FimA Ab attenuated bacterial attachment and aggregation on human gingival and rheumatoid arthritis synovial fibroblasts. In addition, we discovered bacteria may utilize dendritic cells, macrophages and neutrophils to migrate into the joints of CIA mice. These results suggest that disrupting Pg fimbriae function by FimA Ab ameliorates RA.
Collapse
Affiliation(s)
- Sang Hoon Jeong
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Yoojun Nam
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Hyerin Jung
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Juryun Kim
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Yeri Alice Rim
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Narae Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Kijun Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Seungjin Choi
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Yeonsue Jang
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Yena Kim
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ji-Hoi Moon
- Department of Maxillofacial Biomedical Engineering, School of Dentistry, and Department of Life and Nanopharmaceutical Sciences, Kyung Hee University, Seoul, South Korea
| | - Seung Min Jung
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ji Hyeon Ju
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
20
|
Miao P, Zhou XW, Wang P, Zhao R, Chen N, Hu CY, Chen XH, Qian L, Yu QW, Zhang JY, Xu R, He DY, Xiao LB, Li P, Lu M, Zhang DQ. Regulatory effect of anti-gp130 functional mAb on IL-6 mediated RANKL and Wnt5a expression through JAK-STAT3 signaling pathway in FLS. Oncotarget 2018; 9:20366-20376. [PMID: 29755657 PMCID: PMC5945543 DOI: 10.18632/oncotarget.23917] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 12/11/2017] [Indexed: 12/22/2022] Open
Abstract
We investigated the effect on rheumatoid arthritis (RA) of an anti-gp130 monoclonal antibody (mAb) and its mechanism using RA fibroblast-like synoviocytes (FLS) and a collagen antibody–induced arthritis (CAIA) mouse model. We determined the interleukin 6 (IL-6), IL-6 receptor α (IL-6Rα), gp130, receptor activator of nuclear factor κB ligand (RANKL), matrix metalloproteinase 3 (MMP3), TIMP metallopeptidase inhibitor 1 (TIMP1), and Bcl-2 levels in RA and osteoarthritis (OA) serum and synovial fluid. RA FLS were cultured with or without IL-6/IL-6Rα; WNT5A and RANKL levels were detected. We generated an anti-gp130 mAb (M10) with higher affinity and specificity, blocked IL-6 signaling with it, and assessed its effects on the CAIA model, WNT5A and RANKL expression, and signal transducer and activator of transcription 3 (STAT3) phosphorylation. The IL-6 signaling system in patients with RA was increased; RANKL, MMP3, TIMP1, and Bcl-2 in RA bone were elevated. IL-6/IL-6Rα increased RA FLS WNT5A and RANKL expression. M10 ameliorated arthritis in the CAIA model, and inhibited RANKL, WNT5A, and Bcl-2 expression in RA FLS by blocking IL-6 signaling, likely via Janus kinase–STAT3 pathway downregulation. The IL-6–soluble IL-6Rα–gp130 complex is hyperactive in RA and OA. M10 may be the basis for a novel RA treatment drug.
Collapse
Affiliation(s)
- Ping Miao
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Wei Zhou
- Reproductive Medical Center of Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Wang
- Shanghai Jiao Tong University School of Medicine, XinHua Hospital, Shanghai, China.,Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Zhao
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ninan Chen
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Pediatrics, Ruijin Hospital and Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao Ying Hu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Central laboratory, Shanghai Xuhui Central Hospital, Shanghai, China
| | - Xue Hua Chen
- Department of Pediatrics, Ruijin Hospital and Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liu Qian
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Wen Yu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ji Ying Zhang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Xu
- Shanghai Guanghua Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Dong Yi He
- Shanghai Guanghua Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Lian Bo Xiao
- Shanghai Guanghua Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Pu Li
- Department of Pediatrics, Ruijin Hospital and Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Dong Qing Zhang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Ganesan R, Rasool M. Interleukin 17 regulates SHP-2 and IL-17RA/STAT-3 dependent Cyr61, IL-23 and GM-CSF expression and RANKL mediated osteoclastogenesis by fibroblast-like synoviocytes in rheumatoid arthritis. Mol Immunol 2017; 91:134-144. [PMID: 28898718 DOI: 10.1016/j.molimm.2017.09.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/30/2017] [Accepted: 09/01/2017] [Indexed: 01/08/2023]
Abstract
Interleukin (IL)-17 predominately produced by the Th17 cells, plays a crucial role in the fibroblast-like synoviocytes (FLS) mediated disease process of rheumatoid arthritis (RA). IL-17 exerts its pathogenic effects in RA-FLS by IL-17/IL-17RA/STAT-3 signaling. Recent studies have shown that RA-FLS produces SHP-2, Cyr61, IL-23, GM-CSF and RANKL which results in worsening of the disease. However, whether IL-17/IL-17RA/STAT-3 signaling regulates SHP-2, Cyr61, IL-23, GM-CSF and RANKL expressions in RA-FLS remains unknown. In this study, IL-17 treatment dramatically induced the production of Cyr61, IL-23 and GM-CSF in FLS isolated from adjuvant induced arthritis (AA) rats. Conversely, IL-17 mediated production of Cyr61, IL-23 and GM-CSF was abrogated by knockdown of IL-17RA using a small interfering RNA or blockade of STAT-3 activation with S3I-201 in AA-FLS. Interestingly, IL-17 treatment noticeably increased the expression of IL-17RA and SHP-2 in AA-FLS. However, silencing of IL-17RA reversed the effect of IL-17 on the expression of IL-17RA and SHP-2 in AA-FLS. In addition, an increased number of TRAP-positive multinucleated cells were observed in a coculture system consisting of IL-17 treated AA-FLS and rat bone marrow derived monocytes/macrophages. Further, mechanistically we found that IL-17 upregulated RANKL expression in AA-FLS that was dependent on the IL-17/IL-17RA/STAT-3 signaling cascade. Knockdown of IL-17RA or inhibition of STAT-3 activation decreased the IL- 17 induced RANKL expression by AA-FLS and their osteoclastogenic potential. Taken together, our findings demonstrate that IL-17 regulates SHP-2 expression and IL-17RA/STAT-3 dependent production of Cyr61, IL-23, GM-CSF and RANKL in AA-FLS and may reveal a new insight into the pathogenesis of RA.
Collapse
Affiliation(s)
- Ramamoorthi Ganesan
- Immunopathology Lab, School of Bio Sciences and Technology, VIT University, Vellore 632 014, Tamilnadu, India
| | - Mahaboobkhan Rasool
- Immunopathology Lab, School of Bio Sciences and Technology, VIT University, Vellore 632 014, Tamilnadu, India.
| |
Collapse
|
22
|
Nosenko MA, Atretkhany KSN, Mokhonov VV, Efimov GA, Kruglov AA, Tillib SV, Drutskaya MS, Nedospasov SA. VHH-Based Bispecific Antibodies Targeting Cytokine Production. Front Immunol 2017; 8:1073. [PMID: 28919896 PMCID: PMC5585155 DOI: 10.3389/fimmu.2017.01073] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Accepted: 08/17/2017] [Indexed: 12/21/2022] Open
Abstract
Proinflammatory cytokines, such as TNF, IL-6, and IL-1, play pathogenic roles in multiple diseases and are attractive targets for biologic drugs. Because proinflammatory cytokines possess non-redundant protective and immunoregulatory functions, their systemic neutralization carries the potential for unwanted side effects. Therefore, next-generation anti-cytokine therapies would seek to selectively neutralize pathogenic cytokine signaling, leaving normal function intact. Fortunately, the biology of proinflammatory cytokines provides several such opportunities. Here, we discuss various applications of bispecific antibodies targeting cytokines with specific focus on selective TNF neutralization targeted directly to the surface of specific populations of monocytes and macrophages. These bispecific antibodies combine an anti-TNF VHH with VHHs or scFvs directed against abundant surface molecules on myeloid cells and serve to limit the bioavailability of TNF produced by these cells. Such reagents may become prototypes of a novel class of anti-cytokine biologics.
Collapse
Affiliation(s)
- Maxim A. Nosenko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | - Kamar-Sulu N. Atretkhany
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | - Vladislav V. Mokhonov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Grigory A. Efimov
- Lomonosov Moscow State University, Moscow, Russia
- National Research Center for Hematology, Moscow, Russia
| | - Andrey A. Kruglov
- Lomonosov Moscow State University, Moscow, Russia
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- German Rheumatism Research Center, Leibniz Institute, Berlin, Germany
| | - Sergei V. Tillib
- Lomonosov Moscow State University, Moscow, Russia
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Marina S. Drutskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | - Sergei A. Nedospasov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- German Rheumatism Research Center, Leibniz Institute, Berlin, Germany
| |
Collapse
|
23
|
Zhai KF, Duan H, Luo L, Cao WG, Han FK, Shan LL, Fang XM. Protective effects of paeonol on inflammatory response in IL-1β-induced human fibroblast-like synoviocytes and rheumatoid arthritis progression via modulating NF-κB pathway. Inflammopharmacology 2017; 25:10.1007/s10787-017-0385-5. [PMID: 28799079 DOI: 10.1007/s10787-017-0385-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/01/2017] [Indexed: 12/31/2022]
Abstract
Various investigations have demonstrated that human fibroblast-like synoviocytes rheumatoid arthritis (HFLS-RA) take part in the chronic inflammatory responses and RA progression. Inhibition of synovium activation and inflammatory processes may represent a therapeutic target to alleviate RA. Paeonol, a major natural product, has many biological and pharmacological activities. However, its protective effects against RA considering HFLS-RA have not been explored. In this study, anti-inflammatory effects of paeonol were detected in interleukin-1β (IL-1β)-treated HFLS-RA. Our results demonstrated that paeonol had no effect on cell survival and IL-1β-induced proliferation in HFLS-RA. Pretreatment with paeonol significantly suppressed the production of pro-inflammatory TNF-α, IL-6 and IL-1β, and the expressions of matrix metalloproteinase-1/-3 in vitro and in vivo. Mice treated with paeonol (10 mg/kg) remarkablely attenuated arthritic symptoms based on clinical arthritis scores and histopathology in collagen-induced arthritis mice. Furthermore, the TLR4 expression and NF-κB p65 activation were inhibited by paeonol in vitro and in vivo. Our findings illustrated that paeonol had significantly suppressed inflammation effects in synovial tissues and RA progression. The potential mechanism might be based on the attenuation TLR4-NF-κB activation. These collective results indicated that paeonol might be a promising therapeutic agent for alleviating RA progress through inhibiting inflammations and NF-κB signalling pathway.
Collapse
Affiliation(s)
- Ke-Feng Zhai
- Institute of Pharmaceutical Biotechnology, School of Biological and Food Engineering, Suzhou University, 49, Bianhe Road, Suzhou, 234000, People's Republic of China.
- Department of Clinical Laboratory, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, China.
| | - Hong Duan
- Institute of Pharmaceutical Biotechnology, School of Biological and Food Engineering, Suzhou University, 49, Bianhe Road, Suzhou, 234000, People's Republic of China.
| | - Lin Luo
- Institute of Pharmaceutical Biotechnology, School of Biological and Food Engineering, Suzhou University, 49, Bianhe Road, Suzhou, 234000, People's Republic of China
| | - Wen-Gen Cao
- Institute of Pharmaceutical Biotechnology, School of Biological and Food Engineering, Suzhou University, 49, Bianhe Road, Suzhou, 234000, People's Republic of China
| | - Fang-Kai Han
- Institute of Pharmaceutical Biotechnology, School of Biological and Food Engineering, Suzhou University, 49, Bianhe Road, Suzhou, 234000, People's Republic of China
| | - Ling-Ling Shan
- Institute of Pharmaceutical Biotechnology, School of Biological and Food Engineering, Suzhou University, 49, Bianhe Road, Suzhou, 234000, People's Republic of China
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xue-Mei Fang
- Institute of Pharmaceutical Biotechnology, School of Biological and Food Engineering, Suzhou University, 49, Bianhe Road, Suzhou, 234000, People's Republic of China
| |
Collapse
|
24
|
Xu T, Ying T, Wang L, Zhang XD, Wang Y, Kang L, Huang T, Cheng L, Wang L, Zhao Q. A native-like bispecific antibody suppresses the inflammatory cytokine response by simultaneously neutralizing tumor necrosis factor-alpha and interleukin-17A. Oncotarget 2017; 8:81860-81872. [PMID: 29137228 PMCID: PMC5669854 DOI: 10.18632/oncotarget.19899] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 07/12/2017] [Indexed: 01/16/2023] Open
Abstract
Anti-tumor necrosis factor (TNF) therapies are successful in the treatment of inflammatory disorders. However, some patients with rheumatoid arthritis (RA) fail to response anti-TNF drugs due to the compensation of other inflammatory signals. In this study, to reduce compensatory responses of interleukin-17A (IL-17A) during TNF-α inhibition, we generated an IgG-like bispecific antibodiy (bsAb) against TNF-α and IL-17A through a combination method of electrostatic Fc pairing and light chain crossover. This bsAb exhibited relatively high stability comparable to natural IgG antibodies, and retained the unaltered affinities to both of two targets. BsAb significantly decreased not only the expression level of neutrophil or Th17 chemokines, but also the secretion of IL-6/IL-8 on fibroblast-like synoviocytes (FLS) from a patient with RA. Meanwhile, TNF-α-mediated cellular cytotoxicity of fibroblasts was neutralized by bsAb. Importantly, we demonstrate that the combined blockade of TNF-α and IL-17A is more efficient than inhibition of either factor alone. Our results suggest the IgG-like anti-TNF-α/IL-17A bispecific molecule overcome the limited therapeutic responses using anti-TNF drugs. It may be a promising therapeutic agent for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Tianshu Xu
- School of Life Science, Jilin University, Changchun, Jilin, China
| | - Tianlei Ying
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lili Wang
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | | | - Ying Wang
- Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China
| | - Lishan Kang
- Novo Nordisk Research Centre China, Beijing, China
| | - Tao Huang
- Novo Nordisk Research Centre China, Beijing, China
| | - Liang Cheng
- Novo Nordisk Research Centre China, Beijing, China
| | - Liping Wang
- School of Life Science, Jilin University, Changchun, Jilin, China
| | - Qi Zhao
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
25
|
Hollevoet K, Declerck PJ. State of play and clinical prospects of antibody gene transfer. J Transl Med 2017; 15:131. [PMID: 28592330 PMCID: PMC5463339 DOI: 10.1186/s12967-017-1234-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 05/31/2017] [Indexed: 12/31/2022] Open
Abstract
Recombinant monoclonal antibodies (mAbs) are one of today's most successful therapeutic classes in inflammatory diseases and oncology. A wider accessibility and implementation, however, is hampered by the high product cost and prolonged need for frequent administration. The surge in more effective mAb combination therapies further adds to the costs and risk of toxicity. To address these issues, antibody gene transfer seeks to administer to patients the mAb-encoding nucleotide sequence, rather than the mAb protein. This allows the body to produce its own medicine in a cost- and labor-effective manner, for a prolonged period of time. Expressed mAbs can be secreted systemically or locally, depending on the production site. The current review outlines the state of play and clinical prospects of antibody gene transfer, thereby highlighting recent innovations, opportunities and remaining hurdles. Different expression platforms and a multitude of administration sites have been pursued. Viral vector-mediated mAb expression thereby made the most significant strides. Therapeutic proof of concept has been demonstrated in mice and non-human primates, and intramuscular vectored mAb therapy is under clinical evaluation. However, viral vectors face limitations, particularly in terms of immunogenicity. In recent years, naked DNA has gained ground as an alternative. Attained serum mAb titers in mice, however, remain far below those obtained with viral vectors, and robust pharmacokinetic data in larger animals is limited. The broad translatability of DNA-based antibody therapy remains uncertain, despite ongoing evaluation in patients. RNA presents another emerging platform for antibody gene transfer. Early reports in mice show that mRNA may be able to rival with viral vectors in terms of generated serum mAb titers, although expression appears more short-lived. Overall, substantial progress has been made in the clinical translation of antibody gene transfer. While challenges persist, clinical prospects are amplified by ongoing innovations and the versatility of antibody gene transfer. Clinical introduction can be expedited by selecting the platform approach currently best suited for the mAb or disease of interest. Innovations in expression platform, administration and antibody technology are expected to further improve overall safety and efficacy, and unlock the vast clinical potential of antibody gene transfer.
Collapse
Affiliation(s)
- Kevin Hollevoet
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven - University of Leuven, Campus Gasthuisberg O&N 2, P.B. 820, Herestraat 49, 3000 Leuven, Belgium
| | - Paul J. Declerck
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven - University of Leuven, Campus Gasthuisberg O&N 2, P.B. 820, Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
26
|
Wang S, Liang S, Zhao X, He Y, Qi Y. Propofol inhibits cell proliferation and invasion in rheumatoid arthritis fibroblast-like synoviocytes via the nuclear factor-κB pathway. Am J Transl Res 2017; 9:2429-2436. [PMID: 28559993 PMCID: PMC5446525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/25/2017] [Indexed: 06/07/2023]
Abstract
Propofol is an anesthetic drug commonly used in the clinical practice. The aim of this study is to explore the effect of propofol on the aggressive behaviors of rheumatoid arthritis fibroblast-like synoviocytes (RA-FLSs). Propofol treatment for 48 or 72 h significantly inhibited the viability of RA-FLSs, but a 24-h treatment did not produce cytotoxic effects. Propofol exposure for 48 h led to reduction of proliferation and induction of apoptosis in RA-FLSs, which was coupled with increased Bax and decreased Bcl-2 and survivin levels. Additionally, treatment with propofol for 24 h significantly suppressed the migration and invasion of RA-FLSs. Mechanistically, propofol inhibited nuclear factor-κB (NF-κB) activity. Overexpression of constitutively active NF-κB p65 reversed the inhibitory effects of propofol on RA-FLSs. Taken together, propofol exerts anti-proliferative and anti-invasive effects on RA-FLSs via the NF-κB pathway and may have therapeutic potential in treatment of RA.
Collapse
Affiliation(s)
- Song Wang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou UniversityZhengzhou, China
| | - Shuhong Liang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou UniversityZhengzhou, China
| | - Xiaoyu Zhao
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou UniversityZhengzhou, China
| | - Yujie He
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou UniversityZhengzhou, China
| | - Yuedong Qi
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou UniversityZhengzhou, China
| |
Collapse
|
27
|
Wu L, Guo Q, Yang J, Ni B. Tumor Necrosis Factor Alpha Promotes Osteoclast Formation Via PI3K/Akt Pathway-Mediated Blimp1 Expression Upregulation. J Cell Biochem 2017; 118:1308-1315. [PMID: 27505147 DOI: 10.1002/jcb.25672] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 08/08/2016] [Indexed: 01/08/2023]
Abstract
Tumor necrosis factor alpha (TNF-α)-induced osteoclastogenesis have profound effects in states of inflammatory osteolysis such as rheumatoid arthritis, periprosthetic implant loosening, and periodontitis. However, the exact mechanisms by which TNF-α promotes RANKL-induced osteoclast formation remains poorly understood. B lymphocyte-induced maturation protein-1 (Blimp1) is a transcriptional repressor that plays crucial roles in the differentiation and/or function of various kinds of cells including osteoclasts. A novel mechanism was identified where TNF-α-mediated Blimp1 expression, which contributed to RANKL-induced osteoclastogenesis. It is shown that TNF-α could promote the level of Blimp1 expression during osteoclast differentiation. Silencing of Blimp1 in osteoclast precursor cells obviously attenuated the stimulatory effect of TNF-α on osteoclastogenesis. Mechanistically, TNF-α-induced Blimp1 expression was markedly rescued by blocking the PI3K/Akt signaling pathway, which suggested that PI3K/Akt signaling was involved in the regulation of TNF-α-stimulated Blimp1 expression. Taken together, the results established a molecular mechanism of TNF-α-induced osteoclasts differentiation, and provided insights into the potential contribution of Blimp1 in the regulation of osteoclastogenesis by TNF-α. J. Cell. Biochem. 118: 1308-1315, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- LeCheng Wu
- Department of Orthopedics, Changzheng Hospital, The Second Military Medical University, Shanghai, People's Republic of China
| | - QunFeng Guo
- Department of Orthopedics, Changzheng Hospital, The Second Military Medical University, Shanghai, People's Republic of China
| | - Jun Yang
- Department of Orthopedics, Changzheng Hospital, The Second Military Medical University, Shanghai, People's Republic of China
| | - Bin Ni
- Department of Orthopedics, Changzheng Hospital, The Second Military Medical University, Shanghai, People's Republic of China
| |
Collapse
|