1
|
Xiang M, Liu T, Tian C, Ma K, Gou J, Huang R, Li S, Li Q, Xu C, Li L, Lee CH, Zhang Y. Kinsenoside attenuates liver fibro-inflammation by suppressing dendritic cells via the PI3K-AKT-FoxO1 pathway. Pharmacol Res 2022; 177:106092. [PMID: 35066108 PMCID: PMC8776354 DOI: 10.1016/j.phrs.2022.106092] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 12/25/2022]
Abstract
Kinsenoside (KD) exhibits anti-inflammatory and immunosuppressive effects. Dendritic cells (DCs) are critical regulators of the pathologic inflammatory milieu in liver fibrosis (LF). Herein, we explored whether and how KD repressed development of LF via DC regulation and verified the pathway involved in the process. Given our analysis, both KD and adoptive transfer of KD-conditioned DCs conspicuously reduced hepatic histopathological damage, proinflammatory cytokine release and extracellular matrix deposition in CCl4-induced LF mice. Of note, KD restrained the LF-driven rise in CD86, MHC-II, and CCR7 levels and, simultaneously, upregulated PD-L1 expression on DCs specifically, which blocked CD8+T cell activation. Additionally, KD reduced DC glycolysis, maintained DCs immature, accompanied by IL-12 decrease in DCs. Inhibiting DC function by KD disturbed the communication of DCs and HSCs with the expression or secretion of α-SMA and Col-I declined in the liver. Mechanistically, KD suppressed the phosphorylation of PI3K-AKT driven by LF or PI3K agonist, followed by enhanced nuclear transport of FoxO1 and upregulated interaction of FoxO1 with the PD-L1 promoter in DCs. PI3K inhibitor or si-IL-12 acting on DC could relieve LF, HSC activation and diminish the effect of KD. In conclusion, KD suppressed DC maturation with promoted PD-L1 expression via PI3K-AKT-FoxO1 and decreased IL-12 secretion, which blocked activation of CD8+T cells and HSCs, thereby alleviating liver injury and fibro-inflammation in LF.
Collapse
Affiliation(s)
- Ming Xiang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tingting Liu
- Department of Pharmacy, the First Affiliated Hospital of Anhui Medical University, the Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui, China
| | - Cheng Tian
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kun Ma
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jing Gou
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Rongrong Huang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Senlin Li
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qing Li
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chuanrui Xu
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lei Li
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chih-Hao Lee
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, MA, USA
| | - Yonghui Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
2
|
Zhao Y, Sun X, Yang X, Zhang B, Li S, Han P, Zhang B, Wang X, Li S, Chang Y, Wei W. Tolerogenic Dendritic Cells Generated by BAFF Silencing Ameliorate Collagen-Induced Arthritis by Modulating the Th17/Regulatory T Cell Balance. THE JOURNAL OF IMMUNOLOGY 2019; 204:518-530. [PMID: 31843958 DOI: 10.4049/jimmunol.1900552] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/15/2019] [Indexed: 02/06/2023]
Abstract
Tolerogenic dendritic cells (tolDCs) have received much attention because of their capacity to restore immune homeostasis. RNA interference techniques have been used in several studies to generate tolDCs by inactivating certain molecules that regulate DC maturation and immunologic function. BAFF is a key B cell survival factor that is not only essential for B cell function but also T cell costimulation, and DCs are the major source of BAFF. In this study, we determined whether BAFF gene silencing in mature DCs could lead to a tolerogenic phenotype as well as the potential therapeutic effect of BAFF-silenced DCs on collagen-induced arthritis (CIA) in mice. Meanwhile, CRISPR/Cas9-mediated BAFF-/- DC2.4 cells were generated to verify the role of BAFF in DC maturation and functionality. BAFF-silenced DCs and BAFF-/- DC2.4 cells exhibited an immature phenotype and functional state. Further, the transplantation of BAFF-silenced DCs significantly alleviated CIA severity in mice, which correlated with a reduction in Th17 populations and increased regulatory T cells. In vitro, BAFF-silenced DCs promoted Foxp3 mRNA and IL-10 expression but inhibited ROR-γt mRNA and IL-17A expression in CD4+ T cells. Together, BAFF-silenced DCs can alleviate CIA, partly by inducing Foxp3+ regulatory T cells and suppressing Th17 subsets. Collectively, BAFF plays an important role in interactions between DCs and T cells, which might be a promising genetic target to generate tolDCs for autoimmune arthritis treatment.
Collapse
Affiliation(s)
- Yingjie Zhao
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei 230032, China; and
| | - Xiaojing Sun
- Anhui Maternity and Child Health Care Hospital, Hefei 230001, China
| | - Xuezhi Yang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei 230032, China; and
| | - Bingjie Zhang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei 230032, China; and
| | - Siyu Li
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei 230032, China; and
| | - Ping Han
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei 230032, China; and
| | - Binbin Zhang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei 230032, China; and
| | - Xinwei Wang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei 230032, China; and
| | - Susu Li
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei 230032, China; and
| | - Yan Chang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei 230032, China; and
| | - Wei Wei
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei 230032, China; and
| |
Collapse
|
3
|
Suzuki M, Matsumoto T, Toyoda K, Nakamura Y, Murakami S. Impacts of CD40- and CD86-Silenced Antigen-Specific B Cells on the Control of Allergies. Am J Rhinol Allergy 2019; 33:513-523. [PMID: 31064207 DOI: 10.1177/1945892419848188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background We previously reported that CD40-silenced B cells inhibited allergic responses and symptoms. However, more potent therapies are needed. To our knowledge, the effects of CD86-silenced B cells and synergic effects of gene silencing in B cells by 2 small interfering RNAs (siRNAs) on allergic disease control have not been reported. Objective To investigate the effects of CD86-silenced B cells and synergic effects of gene silencing in B cells on allergic responses and symptoms. Methods Mice were treated with CD40- and/or CD86-silenced B cells transfected with siRNAs and pulsed with ovalbumin (OVA). And the effects of these B cells were estimated. Results CD86-silenced OVA-pulsed B cells significantly inhibited OVA-induced allergies. Treatment with CD40-/CD86-silenced OVA-pulsed B cells led to a significantly fewer sneezes and nasal rubbing movements, as well as lower OVA-specific immunoglobulin E (IgE) levels, than that with CD40-silenced or CD86-silenced OVA-pulsed B cells alone. These inhibitory effects were observed prior to sensitization as well as after the establishment of allergic rhinitis. CD40-/CD86-silenced OVA-pulsed B cells did not inhibit keyhole limpet hemocyanin-induced allergies. CD40-/CD86-silenced OVA-pulsed B cells also significantly inhibited allergic symptoms and OVA-specific IgE level in sera compared with CD40-/CD86-silenced OVA-pulsed dendritic cells (DCs). In addition, CD19+CD40− B cells significantly increased in the nasal tissue after intravenous administration of these cells. Furthermore, CD40-/CD86-silenced B cells inhibited allergic symptoms caused by Cry j 1, a major aeroallergen of Japanese cedar pollen, and Cry j 1-specific IgE in sera. Conclusion This study showed, for the first time, that siRNA-induced CD86-silenced B cells significantly inhibited allergic responses and symptoms antigen-specifically, and that siRNA-induced CD40-/CD86-silenced antigen-specific B cells are a more useful antigen-specific therapy than CD40- or CD86-silenced B cells alone for the control of allergies. Furthermore, it was shown that CD40-/CD86-silenced B cells have stronger inhibition of IgE production and allergic symptoms than CD40-/CD86-silenced DCs.
Collapse
Affiliation(s)
- Motohiko Suzuki
- 1 Department of Otorhinolaryngology, Nagoya City University, Nagoya, Japan
| | - Tamami Matsumoto
- 1 Department of Otorhinolaryngology, Nagoya City University, Nagoya, Japan
| | - Kiichi Toyoda
- 1 Department of Otorhinolaryngology, Nagoya City University, Nagoya, Japan
| | - Yoshihisa Nakamura
- 1 Department of Otorhinolaryngology, Nagoya City University, Nagoya, Japan
| | - Shingo Murakami
- 1 Department of Otorhinolaryngology, Nagoya City University, Nagoya, Japan
| |
Collapse
|
4
|
Yang Z, Liu Y, Zhou X. Immune modulation by silencing CD80 and CD86 production in dendritic cells using small hairpin RNA to reduce heart transplant rejection. Transpl Immunol 2018; 49:20-27. [DOI: 10.1016/j.trim.2018.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 03/13/2018] [Accepted: 03/21/2018] [Indexed: 01/30/2023]
|
5
|
TIGIT-Fc alleviates acute graft-versus-host disease by suppressing CTL activation via promoting the generation of immunoregulatory dendritic cells. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3085-3098. [PMID: 29960041 DOI: 10.1016/j.bbadis.2018.06.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 06/25/2018] [Accepted: 06/26/2018] [Indexed: 02/07/2023]
Abstract
Graft-versus-host disease (GVHD) is the most common complication and major limitation of allogeneic hematopoietic stem cell transplantation. The CD226/TIGIT-CD155 signal is critical for the cross-talk between T cells and dendritic cells (DCs). Studies have shown that blockade of the CD226-CD155 interaction, using an anti-CD226 antibody, can significantly ameliorate GVHD. It has also been reported that a TIGIT-Fc fusion protein exerts immunosuppressive effects by binding to CD155 on DCs. Here, we used a mouse allogeneic acute GVHD model to explore the therapeutic potential and mechanism of action of TIGIT-Fc. C57/BL6 and Balb/c mice were used as hematopoietic cell graft donors and recipients, respectively. In the TIGIT-Fc-treated mice, GVHD symptom occurrence and mortality were delayed compared to that in isotype control group mice. Histopathological analyses revealed that following TIGIT-Fc treatment, liver and small intestine tissue damage was reduced with minimal lymphocytic infiltration. The percentage of CD8+IFN-γ+ and CD8+ granzyme B+ cells significantly decreased in the TIGIT-Fc group. Moreover, treatment with TIGIT-Fc, even after the onset of GVHD, ameliorated symptoms and prolonged survival. TIGIT-Fc also inhibited CD8+ T cell activation in vitro; this was dependent on the presence of CD155 on bone marrow-derived dendritic cells (BMDCs) and on IL-10 production. In addition, TIGIT-CD155 ligation triggered both Erk phosphorylation and STAT3 nuclear translocation. These data indicate that TIGIT plays an important role in the development of GVHD and is an ideal molecular target to treat acute GVHD.
Collapse
|
6
|
Zhu Z, Xu R, Zheng X, Wang T, Li D, Wang Y, Liu K. Inhibitory Effect of TLR4 Gene Silencing on Intimal Hyperplasia of Vein Grafting. Vasc Endovascular Surg 2016; 50:464-469. [PMID: 27681173 DOI: 10.1177/1538574416670308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PURPOSE The present study aimed to explore the regulating effect of Toll-like receptor 4 (TLR4) on intimal hyperplasia in rat vein grafts. METHODS Rat models of external jugular vein carotid artery bypass grafting were established. Afterward, TLR4 small interfering RNA (siRNA) recombinant plasmids were constructed, which were transfected into rat vein graft bypass to study the effect of TLR4 silencing on intimal hyperplasia and to explore the underlying mechanisms. Real-time polymerase chain reaction and Western blot were used to detect the expression levels of TLR4 and inflammatory factors in TLR4 siRNA-transfected vein graft bypass. The intimal thickness was evaluated using hematoxylin-eosin staining. RESULTS Compared with the scramble siRNA group, the intimal thickness of vein grafting was decreased significantly, while the inflammatory factors including interleukin (IL) 1β, IL-6, and tumor necrosis factor α in grafted vein were dramatically downregulated in the TLR4 siRNA group. CONCLUSION These results showed that local silencing of TLR4 in the vein grafts could inhibit intimal hyperplasia by downregulating the expression of inflammatory factors in the vein grafts, suggesting that TLR4 can be used as a new target for therapy of vascular intimal hyperplasia.
Collapse
Affiliation(s)
- Zhicheng Zhu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Rihao Xu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Xiaomei Zheng
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Tiance Wang
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Dan Li
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Yong Wang
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Kexiang Liu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| |
Collapse
|
7
|
Arleevskaya MI, Kravtsova OA, Lemerle J, Renaudineau Y, Tsibulkin AP. How Rheumatoid Arthritis Can Result from Provocation of the Immune System by Microorganisms and Viruses. Front Microbiol 2016; 7:1296. [PMID: 27582741 PMCID: PMC4987382 DOI: 10.3389/fmicb.2016.01296] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 08/05/2016] [Indexed: 12/12/2022] Open
Abstract
The pathogenesis of rheumatoid arthritis (RA), similar to development of a majority of inflammatory and autoimmune disorders, is largely due to an inappropriate or inadequate immune response to environmental challenges. Among these challenges, infectious agents are the undisputed leaders. Since the 1870s, an impressive list of microorganisms suspected of provoking RA has formed, and the list is still growing. Although a definite causative link between a specific infectious agent and the disease has not been established, several arguments support such a possibility. First, in the absence of a defined pathogen, the spectrum of triggering agents may include polymicrobial communities or the cumulative effect of several bacterial/viral factors. Second, the range of infectious episodes (i.e., clinical manifestations caused by pathogens) may vary in the process of RA development from preclinical to late-stage disease. Third, infectious agents might not trigger RA in all cases, but trigger it in a certain subset of the cases, or the disease onset may arise from an unfortunate combination of infections along with, for example, psychological stress and/or chronic joint tissue microtrauma. Fourth, genetic differences may have a role in the disease onset. In this review, two aspects of the problem of “microorganisms and RA” are debated. First, is there an acquired immune deficiency and, in turn, susceptibility to infections in RA patients due to the too frequent and too lengthy infections, which at last break the tolerance of self antigens? Or, second, is there a congenital deficiency in tolerance and inflammation control, which may occur even with ordinary infection frequency and duration?
Collapse
Affiliation(s)
- Marina I Arleevskaya
- Central Research Laboratory, Department of Clinical Laboratory Diagnostics, Kazan State Medical Academy Kazan, Russia
| | - Olga A Kravtsova
- Department of Biochemistry and Biotechnology, Kazan Federal University Kazan, Russia
| | - Julie Lemerle
- Laboratory of Immunology and Immunotherapy, CHU Morvan Brest, France
| | - Yves Renaudineau
- Laboratory of Immunology and Immunotherapy, CHU Morvan Brest, France
| | - Anatoly P Tsibulkin
- Central Research Laboratory, Department of Clinical Laboratory Diagnostics, Kazan State Medical Academy Kazan, Russia
| |
Collapse
|
8
|
MENG FANDONG, WANG SHUAI, JIANG YOUHONG, SUI CHENGGUANG. Antitumor effect of dendritic cells transfected with prostate-specific membrane antigen recombinant adenovirus on prostate cancer: An in vitro study. Mol Med Rep 2016; 13:2124-34. [DOI: 10.3892/mmr.2016.4754] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 09/25/2015] [Indexed: 11/06/2022] Open
|
9
|
Raker VK, Domogalla MP, Steinbrink K. Tolerogenic Dendritic Cells for Regulatory T Cell Induction in Man. Front Immunol 2015; 6:569. [PMID: 26617604 PMCID: PMC4638142 DOI: 10.3389/fimmu.2015.00569] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 10/26/2015] [Indexed: 01/01/2023] Open
Abstract
Dendritic cells (DCs) are highly specialized professional antigen-presenting cells that regulate immune responses, maintaining the balance between tolerance and immunity. Mechanisms via which they can promote central and peripheral tolerance include clonal deletion, the inhibition of memory T cell responses, T cell anergy, and induction of regulatory T cells (Tregs). These properties have led to the analysis of human tolerogenic DCs as a therapeutic strategy for the induction or re-establishment of tolerance. In recent years, numerous protocols for the generation of human tolerogenic DCs have been developed and their tolerogenic mechanisms, including induction of Tregs, are relatively well understood. Phase I trials have been conducted in autoimmune disease, with results that emphasize the feasibility and safety of treatments with tolerogenic DCs. Therefore, the scientific rationale for the use of tolerogenic DCs therapy in the fields of transplantation medicine and allergic and autoimmune diseases is strong. This review will give an overview on efforts and protocols to generate human tolerogenic DCs with focus on IL-10-modulated DCs as inducers of Tregs and discuss their clinical applications and challenges faced in further developing this form of immunotherapy.
Collapse
Affiliation(s)
- Verena K Raker
- Department of Dermatology, University Medical Center Mainz, Johannes Gutenberg-University Mainz , Mainz , Germany
| | - Matthias P Domogalla
- Department of Dermatology, University Medical Center Mainz, Johannes Gutenberg-University Mainz , Mainz , Germany
| | - Kerstin Steinbrink
- Department of Dermatology, University Medical Center Mainz, Johannes Gutenberg-University Mainz , Mainz , Germany
| |
Collapse
|
10
|
Brück J, Pascolo S, Fuchs K, Kellerer C, Glocova I, Geisel J, Dengler K, Yazdi AS, Röcken M, Ghoreschi K. Cholesterol Modification of p40-Specific Small Interfering RNA Enables Therapeutic Targeting of Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:2216-23. [PMID: 26232431 DOI: 10.4049/jimmunol.1402989] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 07/01/2015] [Indexed: 11/19/2022]
Abstract
Small interfering RNA (siRNA)-based therapies allow targeted correction of molecular defects in distinct cell populations. Although efficient in multiple cell populations, dendritic cells (DCs) seem to resist siRNA delivery. Using fluorescence labeling and radiolabeling, we show that cholesterol modification enables siRNA uptake by DCs in vitro and in vivo. Delivery of cholesterol-modified p40 siRNA selectively abolished p40 transcription and suppressed TLR-triggered p40 production by DCs. During immunization with peptide in CFA, cholesterol-modified p40 siRNA generated p40-deficient, IL-10-producing DCs that prevented IL-17/Th17 and IFN-γ/Th1 responses. Only cholesterol-modified p40-siRNA established protective immunity against experimental autoimmune encephalomyelitis and suppressed IFN-γ and IL-17 expression by CNS-infiltrating mononuclear cells without inducing regulatory T cells. Because cholesterol-modified siRNA can thus modify selected DC functions in vivo, it is intriguing for targeted immune therapy of allergic, autoimmune, or neoplastic diseases.
Collapse
Affiliation(s)
- Jürgen Brück
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Steve Pascolo
- Department of Oncology, University Hospital Zurich, CH-8044 Zurich, Switzerland
| | - Kerstin Fuchs
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Christina Kellerer
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Ivana Glocova
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Julia Geisel
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Katja Dengler
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Amir S Yazdi
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Martin Röcken
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Kamran Ghoreschi
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| |
Collapse
|
11
|
Versatile polyion complex micelles for peptide and siRNA vectorization to engineer tolerogenic dendritic cells. Eur J Pharm Biopharm 2015; 92:216-27. [DOI: 10.1016/j.ejpb.2015.03.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 03/06/2015] [Accepted: 03/10/2015] [Indexed: 11/20/2022]
|
12
|
Chen S, Tam YYC, Lin PJC, Leung AKK, Tam YK, Cullis PR. Development of lipid nanoparticle formulations of siRNA for hepatocyte gene silencing following subcutaneous administration. J Control Release 2014; 196:106-12. [PMID: 25285610 DOI: 10.1016/j.jconrel.2014.09.025] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Revised: 09/11/2014] [Accepted: 09/25/2014] [Indexed: 02/07/2023]
Abstract
Recently developed lipid nanoparticle (LNP) formulations of siRNA have proven to be effective agents for hepatocyte gene silencing following intravenous administration with at least three LNP-siRNA formulations in clinical trials. The aim of this work was to develop LNP-siRNA systems for hepatocyte gene silencing that can be administered subcutaneously (s.c.). Three parameters were investigated, namely LNP size, residence time of the polyethylene glycol (PEG)-lipid coating and the influence of hepatocyte-specific targeting ligands. LNP sizes were varied over the range of 30 to 115 nm in diameter and PEG-lipid that dissociates rapidly (PEG-DMG) and slowly (PEG-DSG) were employed. In mice, results show that large (~80 nm) LNP exhibited limited accumulation in the liver and poor Factor VII (FVII) gene silencing at 1mg siRNA/kg body weight. Conversely, small (~30 nm) LNP systems showed maximal liver accumulation yet still had minimal activity. Interestingly, intermediate size (~45 nm) LNP containing PEG-DSG exhibited nearly equivalent liver accumulation as the smaller systems following s.c. administration but reduced FVII levels by 80% at 1mg siRNA/kg body weight. Smaller systems (~35 nm diameter) containing either PEG-DMG or PEG-DSG were less active; however addition of 0.5 mol.% of a GalNAc-PEG lipid to these smaller systems improved activity to levels similar to that observed for the ~45 nm diameter systems. In summary, this work shows that appropriately designed LNP-siRNA systems can result in effective hepatocyte gene silencing following s.c administration.
Collapse
Affiliation(s)
- Sam Chen
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia, Canada, V6T 1Z3
| | - Yuen Yi C Tam
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia, Canada, V6T 1Z3
| | - Paulo J C Lin
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia, Canada, V6T 1Z3
| | - Alex K K Leung
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia, Canada, V6T 1Z3
| | - Ying K Tam
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia, Canada, V6T 1Z3; Acuitas Therapeutics, 2714 West 31st Avenue, Vancouver, British Columbia, Canada, V6L 2A1
| | - Pieter R Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia, Canada, V6T 1Z3.
| |
Collapse
|
13
|
Zhang X, Liu Y, Zhang G, Shi J, Zhang X, Zheng X, Jiang AT, Zhang ZX, Johnston N, Siu KS, Chen R, Lian D, Koos D, Quan D, Min WP. Synergic silencing of costimulatory molecules prevents cardiac allograft rejection. J Transl Med 2014; 12:142. [PMID: 24886282 PMCID: PMC4040111 DOI: 10.1186/1479-5876-12-142] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 04/28/2014] [Indexed: 12/13/2022] Open
Abstract
Background While substantial progress has been made in blocking acute transplant rejection with the advent of immune suppressive drugs, chronic rejection, mediated primarily by recipient antigen presentation, remains a formidable problem in clinical transplantation. We hypothesized that blocking co-stimulatory pathways in the recipient by induction of RNA interference using small interference RNA (siRNA) expression vectors can prolong allogeneic heart graft survival. Method Vectors expressing siRNA specifically targeting CD40 and CD80 were prepared. Recipients (BALB/c mice) were treated with CD40 and/or CD80 siRNA expression vectors via hydrodynamic injection. Control groups were injected with a scrambled siRNA vector and sham treatment (PBS). After treatment, a fully MHC-mismatched (BALB/c to C57/BL6) heart transplantation was performed. Result Allogeneic heart graft survival (>100 days) was approximately 70% in the mice treated simultaneously with CD40 and CD80 siRNA expression vectors with overall reduction in lymphocyte interstitium infiltration, vascular obstruction, and edema. Hearts transplanted into CD40 or CD80 siRNA vector-treated recipients had an increased graft survival time compared to negative control groups, but did not survive longer than 40 days. In contrast, allogenic hearts transplanted into recipients treated with scrambled siRNA vector and PBS stopped beating within 10–16 days. Real-time PCR (RT-PCR) and flow cytometric analysis showed an upregulation of FoxP3 expression in spleen lymphocytes and a concurrent downregulation of CD40 and CD80 expression in splenic dendritic cells of siRNA-treated mice. Functional suppressive activity of splenic dendritic cells (DCs) isolated from tolerant recipients was demonstrated in a mixed lymphocyte reaction (MLR). Furthermore, DCs isolated from CD40- and CD80-treated recipients promoted CD4 + CD25 + FoxP3+ regulatory T cell differentiation in vitro. Conclusion This study demonstrates that the simultaneous silencing of CD40 and CD80 genes has synergistic effects in preventing allograft rejection, and may therefore have therapeutic potential in clinical transplantation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Wei-Ping Min
- Department of Surgery, Pathology, and Ocology, University of Western Ontario, London, Canada.
| |
Collapse
|
14
|
Gong X, Han B, Zou Y, Wang J, Yang W. Attenuation of Experimental Autoimmune Myocarditis by si-RNA Mediated CD40 Silencing. Int Heart J 2014; 55:539-45. [DOI: 10.1536/ihj.14-125] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Xin Gong
- Division of Cardiology, Department of Pediatrics, Provincial Hospital Affiliated with Shandong University
- Division of Cardiology, Yantai Yuhuangding Hospital
| | - Bo Han
- Division of Cardiology, Department of Pediatrics, Provincial Hospital Affiliated with Shandong University
| | - Yong Zou
- Department of Combined Traditional Chinese and Western Medicine
| | - Jiezhong Wang
- Division of Cardiology, Department of Pediatrics, Provincial Hospital Affiliated with Shandong University
| | - Wenwei Yang
- Division of Cardiology, Department of Pediatrics, Provincial Hospital Affiliated with Shandong University
| |
Collapse
|
15
|
Siegert I, Schatz V, Prechtel AT, Steinkasserer A, Bogdan C, Jantsch J. Electroporation of siRNA into mouse bone marrow-derived macrophages and dendritic cells. Methods Mol Biol 2014; 1121:111-9. [PMID: 24510816 DOI: 10.1007/978-1-4614-9632-8_9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Dendritic cells (DC) and macrophages (MΦ) play a pivotal role in antimicrobial defense, in the regulation of immune responses, and in maintaining tissue homeostasis. The analysis of DC and MΦ function relies on primary cells albeit these cells are known to be difficult to transfect. This makes the use of small interfering RNA (siRNA) for targeted manipulation of gene expression by RNA interference difficult. In the following chapter, we provide a detailed protocol for the successful transfer of siRNA via electroporation into a defined population of mouse bone marrow-derived MΦ or DC that does not cause toxicity to the myeloid cells or nonspecific alterations of their biological functions. Factors that influence the transfection and knockdown rate will be highlighted.
Collapse
Affiliation(s)
- Isabel Siegert
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | | | | | |
Collapse
|
16
|
Tsuruta T, Inoue R, Nagino T, Nishibayashi R, Makioka Y, Ushida K. Role of the mannose receptor in phagocytosis of Enterococcus faecalis strain EC-12 by antigen-presenting cells. Microbiologyopen 2013; 2:610-7. [PMID: 23801521 PMCID: PMC3831625 DOI: 10.1002/mbo3.99] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 05/20/2013] [Accepted: 05/21/2013] [Indexed: 01/21/2023] Open
Abstract
The aim of this study was to clarify the phagocytic mechanisms of a heat-killed cell preparation of Enterococcus faecalis strain EC-12 (EC-12) by antigen-presenting cells (APCs). Fluorescein isothiocyanate (FITC)-labeled EC-12 was cocultured with peritoneal macrophage and the amount of EC-12 phagocytosed by peritoneal macrophages was measured using a microplate fluorometer. Peritoneal macrophages from toll-like receptor (TLR)2-, TLR7-, and MyD88-deficient knockout (KO) mice exhibited similar levels of EC-12 phagocytosis to those from wild-type mice. Similarly, dectin-1 neutralization of peritoneal macrophages had no effect on EC-12 phagocytosis. However, blockade of the mannose receptor (MR) significantly decreased the amount of EC-12 phagocytosed by peritoneal macrophages; the same effect was observed in bone marrow-derived macrophages and dendritic cells. Our findings suggest that MR plays a major role in EC-12 phagocytosis by the APCs. This aim of this study was to clarify the phagocytic mechanisms of a heat-killed cell preparation of Enterococcus faecalis strain EC-12 (EC-12) by antigen-presenting cells (APCs). Our findings suggest that mannose receptor (MR) plays a major role in EC-12 phagocytosis by the APCs.
Collapse
Affiliation(s)
- Takeshi Tsuruta
- Laboratory of Animal Science, Kyoto Prefectural University, 1-5 Shimogamohangityou, Sakyo-ku, Kyoto, Japan
| | | | | | | | | | | |
Collapse
|
17
|
Zhang X, Beduhn M, Zheng X, Lian D, Chen D, Li R, Siu LKS, Marleau A, French PW, Ichim TE, Min WP. Induction of alloimmune tolerance in heart transplantation through gene silencing of TLR adaptors. Am J Transplant 2012; 12:2675-88. [PMID: 22823145 DOI: 10.1111/j.1600-6143.2012.04196.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Toll-like receptors (TLRs) activate biochemical pathways that evoke activation of innate immunity, which leads to dendritic cell (DC) maturation and initiation of adaptive immune responses that provoke allograft rejection. We aimed to prolong allograft survival by selectively inhibiting expression of the common adaptors of TLR signaling, namely MyD88 and TRIF, using siRNA. In vitro we demonstrated that blocking expression of MyD88 and TRIF led to reduced DC maturation. In vivo treatment of recipients with MyD88 and TRIF siRNA significantly prolonged allograft survival in the BALB/c > C57BL6 cardiac transplant model. Moreover, the combination of MyD88 and TRIF siRNA along with a low dose of rapamycin further extended the allograft survival (88.8 ± 7.1 days). Tissue histopathology demonstrated an overall reduction in lymphocyte interstitium infiltration, vascular obstruction and hemorrhage in mice treated with MyD88 and TRIF siRNA vector plus rapamycin. Furthermore, treatment was associated with an increase in the numbers of CD4(+) CD25(+) FoxP3(+) regulatory T cells and Th2 deviation. To our knowledge, this study is the first demonstration of prolonging the survival of allogeneic heart grafts through gene silencing of TLR signaling adaptors, highlighting the therapeutic potential of siRNA in clinical transplantation.
Collapse
Affiliation(s)
- X Zhang
- Departments of Surgery, Pathology, and Oncology, University of Western Ontario, London, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Zheng X, Koropatnick J, Chen D, Velenosi T, Ling H, Zhang X, Jiang N, Navarro B, Ichim TE, Urquhart B, Min W. Silencing IDO in dendritic cells: a novel approach to enhance cancer immunotherapy in a murine breast cancer model. Int J Cancer 2012; 132:967-77. [PMID: 22870862 DOI: 10.1002/ijc.27710] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 06/14/2012] [Indexed: 12/15/2022]
Abstract
Cancer immunotherapeutic agents (vaccines) in the form of antigen-loaded dendritic cells (DCs) reached an important milestone with the recent approval of Provenge, the first DC vaccine for treatment of prostate cancer. Although this heralds a new era of tumor immunotherapy, it also highlights the compelling need to optimize such DC-based therapies as they are increasingly tested and used to treat human patients. In this study we sought to augment and enhance the antitumor activity of a DC-based vaccine using siRNA to silence expression of immunosuppressive enzyme indoleamine 2,3-dioxygenase (IDO) in DCs. We report here that DCs loaded with tumor antigens, but with siRNA-silenced IDO expression, were introduced into 4T1 breast tumor-bearing mice, the treatment: (i) lengthened the time required for tumor onset, (ii) decreased tumor size compared to tumors grown for equal lengths of time in mice treated with antigen-loaded DCs without IDO silencing and (iii) reduced CD4(+) and CD8(+) T cell apoptosis. Furthermore, immunization with IDO-silenced DCs enhanced tumor antigen-specific T cell proliferation and CTL activity, and decreased numbers of CD4(+) CD25(+) Foxp3(+) T(reg). This study provides evidence to support silencing of immunosuppressive genes (IDO) as an effective strategy to enhance the efficacy of DC-based cancer immunotherapeutic.
Collapse
Affiliation(s)
- Xiufen Zheng
- Department of Surgery, University of Western Ontario, London, Ontario, Canada.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Li R, Zheng X, Popov I, Zhang X, Wang H, Suzuki M, Necochea-Campion RD, French PW, Chen D, Siu L, Koos D, Inman RD, Min WP. Gene silencing of IL-12 in dendritic cells inhibits autoimmune arthritis. J Transl Med 2012; 10:19. [PMID: 22289162 PMCID: PMC3293054 DOI: 10.1186/1479-5876-10-19] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 01/31/2012] [Indexed: 11/18/2022] Open
Abstract
Background We have previously demonstrated that immune modulation can be accomplished by administration of gene silenced dendritic cells (DC) using siRNA. In this study, we demonstrate the therapeutic utilization of shRNA-modified DC as an antigen-specific tolerogenic vaccine strategy for autoimmune arthritis. Methods A shRNA that specifically targets IL-12 p35 was designed and cloned into a plasmid vectors (IL-12 shRNA). Bone marrow-derived DC from DBA/1 mice were transfected with the IL-12 shRNA construct in vitro. Mice with collagen II (CII)-induced arthritis (CIA) were treated with the modified DCs expressing the shRNA. Recall response and disease progression were assessed. Results After gene silencing of IL-12 in DC, DC were shown to selectively inhibit T cell proliferation on recall responses and in an MLR. In murine CIA, we demonstrated that administration of IL-12 shRNA-expressing DC that were pulsed with CII inhibited progression of arthritis. The therapeutic effects were evidenced by decreased clinical scores, inhibition of inflammatory cell infiltration in the joint, and suppression of T cell and B cell responses to CII. Conclusion We demonstrate a novel tolerance-inducing protocol for the treatment of autoimmune inflammatory joint disease in which the target antigen is known, utilizing DNA-directed RNA interference.
Collapse
Affiliation(s)
- Rong Li
- Institute of Immunomodulation and Immunotherapy, Nanchang University Medical School, Nanchang, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Miah MA, Yoon CH, Kim J, Jang J, Seong YR, Bae YS. CISH is induced during DC development and regulates DC-mediated CTL activation. Eur J Immunol 2011; 42:58-68. [PMID: 22002016 DOI: 10.1002/eji.201141846] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 09/16/2011] [Accepted: 10/06/2011] [Indexed: 11/07/2022]
Abstract
The cytokine inducible SH2-domain protein (CISH) is a well-known STAT5 target gene, but its role in the immune system remains uncertain. In this study, we found that CISH is predominantly induced during dendritic cell (DC) development from mouse bone marrow (BM) cells and plays a crucial role in type 1 DC development and DC-mediated CTL activation. CISH knockdown reduced the expression of MHC class I, co-stimulatory molecules and pro-inflammatory cytokines in BMDCs. Meanwhile, the DC yield was markedly enhanced by CISH knockdown via cell-cycle activation and reduction of cell apoptosis. Down-regulation of cell proliferation at the later stage of DC development was found to be associated with CISH-mediated negative feedback regulation of STAT5 activation. In T-cell immunity, OT-1 T-cell proliferation was significantly reduced by CISH knockdown in DCs, whereas OT-2 T-cell proliferation was not affected by CISH knockdown. CTLs generated by DC vaccination were also markedly reduced by CISH knockdown, followed by significant impairment of DC-based tumor immunotherapy. Taken together, our data suggest that CISH expression at the later stage of DC development triggers the shutdown of DC progenitor cell proliferation and facilitates DC differentiation into a potent stimulator of CTLs.
Collapse
Affiliation(s)
- Mohammad Alam Miah
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Gyeonggi-do, South Korea
| | | | | | | | | | | |
Collapse
|
21
|
Jiang W. Blockade of B7-H1 enhances dendritic cell-mediated T cell response and antiviral immunity in HBV transgenic mice. Vaccine 2011; 30:758-66. [PMID: 22133510 DOI: 10.1016/j.vaccine.2011.11.076] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2011] [Revised: 11/01/2011] [Accepted: 11/18/2011] [Indexed: 01/12/2023]
Abstract
Dendritic cells (DCs) have been identified as the most effective antigen-presenting cell (APC), much attention has been directed toward the use of DCs in vaccine strategies for the treatment of cancer or chronic virus infection. B7-H1 (PD-L1) is a ligand for programmed cell death-1 (PD-1) and does not bind to other CD28 family members, it is expressed on resting and upregulated on activated B, T, myeloid, and dendritic cells (DCs). The overwhelming number of studies supports the role of B7-H1 as a negative regulator of T cell responses. RNA interference (RNAi) is a mechanism for sequence-specific posttranscriptional inhibition of gene expression via double stranded RNA molecules and it has recently been applied to mammalian cells with the use of small interfering RNAs (siRNAs). In the study, transfection of DCs with siRNA specific for B7-H1 gene resulted in the blockade of the expression of B7-H1 on DCs. The allostimulatory activity of DCs could be enhanced by silencing of B7-H1 on DCs. Blockade of B7-H1 on DCs inhibited the production of IFN-γ and IL-10 but not IL-2 and IL-4 in mixed lymphocyte reaction (MLR). HBV specific peptide-pulsed DCs could break tolerance and trigger specific CTL responses, the level of HBsAg and HBV DNA in sera of HBV transgenic mice decreased, whereas blockade of B7-H1 on DCs augmented the effects. These data strongly support the concept that blockade of B7-H1 can enhance DC-mediated antiviral immune responses.
Collapse
Affiliation(s)
- Wenzheng Jiang
- School of Life Science, East China Normal University, Shanghai 200062, China.
| |
Collapse
|
22
|
Basha G, Novobrantseva TI, Rosin N, Tam YYC, Hafez IM, Wong MK, Sugo T, Ruda VM, Qin J, Klebanov B, Ciufolini M, Akinc A, Tam YK, Hope MJ, Cullis PR. Influence of cationic lipid composition on gene silencing properties of lipid nanoparticle formulations of siRNA in antigen-presenting cells. Mol Ther 2011; 19:2186-200. [PMID: 21971424 PMCID: PMC3242662 DOI: 10.1038/mt.2011.190] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Lipid nanoparticles (LNPs) are currently the most effective in vivo delivery systems for silencing target genes in hepatocytes employing small interfering RNA. Antigen-presenting cells (APCs) are also potential targets for LNP siRNA. We examined the uptake, intracellular trafficking, and gene silencing potency in primary bone marrow macrophages (bmMΦ) and dendritic cells of siRNA formulated in LNPs containing four different ionizable cationic lipids namely DLinDAP, DLinDMA, DLinK-DMA, and DLinKC2-DMA. LNPs containing DLinKC2-DMA were the most potent formulations as determined by their ability to inhibit the production of GAPDH target protein. Also, LNPs containing DLinKC2-DMA were the most potent intracellular delivery agents as indicated by confocal studies of endosomal versus cytoplamic siRNA location using fluorescently labeled siRNA. DLinK-DMA and DLinKC2-DMA formulations exhibited improved gene silencing potencies relative to DLinDMA but were less toxic. In vivo results showed that LNP siRNA systems containing DLinKC2-DMA are effective agents for silencing GAPDH in APCs in the spleen and peritoneal cavity following systemic administration. Gene silencing in APCs was RNAi mediated and the use of larger LNPs resulted in substantially reduced hepatocyte silencing, while similar efficacy was maintained in APCs. These results are discussed with regard to the potential of LNP siRNA formulations to treat immunologically mediated diseases.
Collapse
Affiliation(s)
- Genc Basha
- NanoMedicine Research Group, Department of Biochemistry and Molecular Biology Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Doffek K, Chen X, Sugg SL, Shilyansky J. Phosphatidylserine inhibits NFκB and p38 MAPK activation in human monocyte derived dendritic cells. Mol Immunol 2011; 48:1771-7. [PMID: 21628073 DOI: 10.1016/j.molimm.2011.04.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 04/05/2011] [Accepted: 04/25/2011] [Indexed: 01/01/2023]
Abstract
Phosphatidylserine (PS) is an anionic phospholipid restricted to the inner surface of the plasma membrane. PS translocates to the cell surface during early apoptosis where it serves as a marker for rapid uptake by phagocytes. PS is also thought to regulate immune responses. Dendritic cells (DC) are the most potent antigen presenting cells. Previous studies demonstrated that PS inhibits the expression of MHC and co-stimulatory molecules, the secretion of IL-12p70, and the ability to activate T cells by human monocyte derived DCs. However, the cell signaling mechanisms by which PS regulated DCs are not well described. In the current study we tested the effects of PS on signal transduction pathways thought to regulate human myeloid DC maturation and IL-12p70 production. We showed that PS inhibited the activation of nuclear factor-κB (NFκB) in response to LPS by preventing IκBα phosphorylation and degradation. PS also increased the total IκBα levels in immature DCs and inhibited p38 mitogen activated protein kinase (MAPK) phosphorylation and activation. The findings suggest a possible mechanism for regulating the immunostimulatory function of DCs by PS.
Collapse
Affiliation(s)
- Kara Doffek
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | | | | | | |
Collapse
|
24
|
Yang B, Elias JE, Bloxham M, Nicholson ML. Synthetic small interfering RNA down-regulates caspase-3 and affects apoptosis, IL-1β, and viability of porcine proximal tubular cells. J Cell Biochem 2011; 112:1337-47. [DOI: 10.1002/jcb.23050] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
25
|
Abstract
Dendritic cells (DCs) play a pivotal role in regulating the balance between immunity and tolerance of the immune system. Recent advancements in DC biology and techniques for manipulating the function of these cells have shown their immense therapeutic potential for treating a variety of immune disorders. Theoretically, antigen-specific tolerogenic DCs can be generated in vitro and delivered to patients to correct the dysfunctional immune responses that attack their own tissues or over-react to innocuous foreign antigens. However, DCs are a heterogeneous population of cells with differences in cell surface makers, differentiation pathways and functions. Studies are needed to examine which subset of DCs can be used for what type of applications. Furthermore, most of the information on tolerogenic DCs has been obtained from animal models and translational studies are needed to examine how a DC therapeutic strategy can be implemented clinically to modulate immunity.
Collapse
Affiliation(s)
- Jim Hu
- Physiology and Experimental Medicine Research Program, Hospital for Sick Children, 555 University Avenue, Toronto, ON, Canada.
| | | |
Collapse
|
26
|
Li F, Mahato RI. RNA interference for improving the outcome of islet transplantation. Adv Drug Deliv Rev 2011; 63:47-68. [PMID: 21156190 DOI: 10.1016/j.addr.2010.11.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 11/19/2010] [Accepted: 11/25/2010] [Indexed: 01/06/2023]
Abstract
Islet transplantation has the potential to cure type 1 diabetes. Despite recent therapeutic success, it is still not common because a large number of transplanted islets get damaged by multiple challenges including instant blood mediated inflammatory reaction, hypoxia/reperfusion injury, inflammatory cytokines, and immune rejection. RNA interference (RNAi) is a novel strategy to selectively degrade target mRNA. The use of RNAi technologies to downregulate the expression of harmful genes has the potential to improve the outcome of islet transplantation. The aim of this review is to gain a thorough understanding of biological obstacles to islet transplantation and discuss how to overcome these barriers using different RNAi technologies. This eventually will help improve islet survival and function post transplantation. Chemically synthesized small interferring RNA (siRNA), vector based short hairpin RNA (shRNA), and their critical design elements (such as sequences, promoters, and backbone) are discussed. The application of combinatorial RNAi in islet transplantation is also discussed. Last but not the least, several delivery strategies for enhanced gene silencing are discussed, including chemical modification of siRNA, complex formation, bioconjugation, and viral vectors.
Collapse
Affiliation(s)
- Feng Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | | |
Collapse
|
27
|
Zhang Y, Peng T, Zhu H, Zheng X, Zhang X, Jiang N, Cheng X, Lai X, Shunnar A, Singh M, Riordan N, Bogin V, Tong N, Min WP. Prevention of hyperglycemia-induced myocardial apoptosis by gene silencing of Toll-like receptor-4. J Transl Med 2010; 8:133. [PMID: 21159162 PMCID: PMC3020152 DOI: 10.1186/1479-5876-8-133] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 12/15/2010] [Indexed: 02/05/2023] Open
Abstract
Background Apoptosis is an early event involved in cardiomyopathy associated with diabetes mellitus. Toll-like receptor (TLR) signaling triggers cell apoptosis through multiple mechanisms. Up-regulation of TLR4 expression has been shown in diabetic mice. This study aimed to delineate the role of TLR4 in myocardial apoptosis, and to block this process through gene silencing of TLR4 in the myocardia of diabetic mice. Methods Diabetes was induced in C57/BL6 mice by the injection of streptozotocin. Diabetic mice were treated with 50 μg of TLR4 siRNA or scrambled siRNA as control. Myocardial apoptosis was determined by TUNEL assay. Results After 7 days of hyperglycemia, the level of TLR4 mRNA in myocardial tissue was significantly elevated. Treatment of TLR4 siRNA knocked down gene expression as well as diminished its elevation in diabetic mice. Apoptosis was evident in cardiac tissues of diabetic mice as detected by a TUNEL assay. In contrast, treatment with TLR4 siRNA minimized apoptosis in myocardial tissues. Mechanistically, caspase-3 activation was significantly inhibited in mice that were treated with TLR4 siRNA, but not in mice treated with control siRNA. Additionally, gene silencing of TLR4 resulted in suppression of apoptotic cascades, such as Fas and caspase-3 gene expression. TLR4 deficiency resulted in inhibition of reactive oxygen species (ROS) production and NADPH oxidase activity, suggesting suppression of hyperglycemia-induced apoptosis by TLR4 is associated with attenuation of oxidative stress to the cardiomyocytes. Conclusions In summary, we present novel evidence that TLR4 plays a critical role in cardiac apoptosis. This is the first demonstration of the prevention of cardiac apoptosis in diabetic mice through silencing of the TLR4 gene.
Collapse
Affiliation(s)
- Yuwei Zhang
- Department of Endocrinology, West China Hospital of Sichuan University, Chengdu, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Karimi MH, Ebadi P, Pourfathollah AA, Moazzeni M, Soheili ZS, Samiee S. Comparison of three techniques for generation of tolerogenic dendritic cells: siRNA, oligonucleotide antisense, and antibody blocking. Hybridoma (Larchmt) 2010; 29:473-80. [PMID: 21087095 DOI: 10.1089/hyb.2010.0060] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
In recent years, a new view of dendritic cells (DCs) as a main regulator of immunity to induce and maintain tolerance has been established. In vitro manipulation of their development and maturation is a topic of DC therapeutic application, which utilizes their inherent tolerogenicity. In this field, the therapeutic potential of antisense, siRNA, and blocking antibody are an interesting goal. In the present study, the efficiency of these three methods--siRNA, antisense, and blocking antibody--against CD40 molecule and its function in DCs and BCL1 cell line are compared. DCs were separated from mouse spleen and then cultured in vitro using Lipofectamine 2000 to deliver both silencers; the efficacy of transfection was estimated by flow cytometry. mRNA expression and protein synthesis were assessed by real time-PCR and flow cytometry, respectively. By Annexin V and propidium iodine staining, we could evaluate the viability of transfected cells. Knocking down the CD40 gene into separate groups of DCs by siRNA, antisense, and blocking antibody treated DCs can cause an increase in IL-4, decrease in IL-12, IFN-γ production, and allostimulation activity. Our results indicated that, in comparison to antisense and blocking antibody, siRNAs appear to be quantitatively more efficient in CD40 downregulation and their differences are significant.
Collapse
|
29
|
A liposome-based platform, VacciMax, and its modified water-free platform DepoVax enhance efficacy of in vivo nucleic acid delivery. Vaccine 2010; 28:6176-82. [PMID: 20656034 DOI: 10.1016/j.vaccine.2010.07.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 06/10/2010] [Accepted: 07/08/2010] [Indexed: 11/24/2022]
Abstract
Nucleic acid vaccines represent a promising alternative to killed bacterial antigen, recombinant protein or peptide vaccines for infectious diseases and cancer immunotherapy. Although significant advances are made with DNA vaccines in animal studies, there are severe limitations to deliver these vaccines effectively and considerable reservations exist about current methods used. In this study, a liposome-based vaccine platform, VacciMax (VM), and its modified water-free version, DepoVax (DPX), were tested for their ability to improve in vivo delivery of plasmid DNA (pDNA), mRNA and siRNA. Subcutaneously injected pDNA for IL12 and pDNA as well as mRNA for green fluorescent protein (GFP) in VM/DPX significantly enhanced their in vivo expression. Enhanced IL12 secretion and GFP expression was restricted to CD11b(+) and CD11c(+) antigen-presenting cells, but not B cells. Further, significant inhibition of plasmid/antigen-induced IL12 secretion was seen after injection of IL12-siRNA in VM. These findings suggest VM and DPX to be promising means of delivering nucleic acid vaccines in vivo, and warrant further studies on their role in inducing effective immune responses.
Collapse
|
30
|
Abe M, Metes D, Thomson AW. Dendritic cells and regulation of alloimmune responses: relevance to outcome and therapy of organ transplantation. Expert Rev Clin Immunol 2010; 1:419-30. [PMID: 20476992 DOI: 10.1586/1744666x.1.3.419] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Dendritic cells are uniquely well-equipped for antigen capture, processing and presentation. They are highly-efficient antigen-presenting cells that induce and regulate T-cell reactivity. Due to their inherent tolerogenicity, immature dendritic cells offer considerable potential as candidate cellular vaccines for negative regulation of immune reactivity/promotion of tolerance. Both classic myeloid and, more recently, characterized plasmacytoid dendritic cells, exhibit tolerogenic properties. Manipulation of dendritic cells differentiation/ maturation in the laboratory using cytokines, pharmacologic agents or genetic engineering approaches can render stably immature dendritic cells that promote organ transplant tolerance in rodents. There are also indications from human studies of the ability of dendritic cells to promote T-cell tolerance and induce T-regulatory cells, with potential for therapeutic application in organ transplantation. In addition, recent clinical observations suggest that modulation of dendritic cell function (e.g., by immunosuppressive drugs) affects the outcome of transplantation. The challenge confronting applied dendritic cell biology is the identification of optimal strategies and therapeutic regimens to allow the potential of these powerful immune regulatory cells to be realized in the clinic.
Collapse
Affiliation(s)
- Masanori Abe
- Thomas E Starzl Transplantation Institute and Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA.
| | | | | |
Collapse
|
31
|
Endoplasmic reticulum stress-mediated apoptosis involved in indirect recognition pathway blockade induces long-term heart allograft survival. J Biomed Biotechnol 2010; 2010:705431. [PMID: 20490280 PMCID: PMC2871569 DOI: 10.1155/2010/705431] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2009] [Accepted: 02/24/2010] [Indexed: 12/30/2022] Open
Abstract
Implementation of dendritic cell- (DC-) based therapies in organ transplantation can reduce dependency on nonspecific immunosuppression. Despite extensive research, mechanisms of equipped DCs inducing transplant tolerance remain incomplete. Here, we applied RNA interference technique to inhibit CD80 and CD86 expression in host bone marrow-derived DCs. This approach could specifically and effectively knock down CD80 and CD86 expression. T cells primed by these DCs inhibited allogeneic responses. Administration of recipient DCs loaded with alloantigen after CD80 and CD86 blockade prolonged cardiac allograft survival. We also found a higher percentage of apoptotic T cells in lymph tissues and grafts than that detected in control group. In addition, these T cells expressed high expression of GRP78 than controls, indicating activation of unfolded protein responses. Upregulation of CHOP expression among these cells suggested that the endoplasmic reticulum stress (ERS) response switched to a proapoptotic response. Our results indicated that ERS-induced apoptosis may be involved in allogeneic T-cell apoptosis, and the ERS-mediated apoptosis pathway may be a novel target in clinical prevention and therapy of allograft rejection.
Collapse
|
32
|
Zheng X, Suzuki M, Ichim TE, Zhang X, Sun H, Zhu F, Shunnar A, Garcia B, Inman RD, Min W. Treatment of autoimmune arthritis using RNA interference-modulated dendritic cells. THE JOURNAL OF IMMUNOLOGY 2010; 184:6457-64. [PMID: 20435931 DOI: 10.4049/jimmunol.0901717] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Dendritic cells (DCs) have a dual ability to either stimulate or suppress immunity, which is primarily associated with the expression of costimulatory molecules. Ag-loaded DCs have shown encouraging clinical results for treating cancer and infectious diseases; however, the use of these cells as a means of suppressing immune responses is only recently being explored. Here, we describe the induction of RNA interference through administering short interfering RNA (siRNA) as a means of specifically generating tolerogenic DCs. Knockdown of CD40, CD80, and CD86, prior to loading DCs with the arthritogenic Ag collagen II, led to a population of cells that could effectively suppress onset of collagen-induced arthritis. Maximum benefits were observed when all three genes were concurrently silenced. Disease suppression was associated with inhibition of collagen II-specific Ab production and suppression of T cell recall responses. Downregulation of IL-2, IFN-gamma, TNF-alpha, and IL-17 and increased FoxP3(+) cells with regulatory activity were observed in collagen-induced arthritis mice treated with siRNA-transfected DCs. Collectively, these data support the use of ex vivo gene manipulation in DCs using siRNA to generate tailor-made tolerogenic vaccines for treating autoimmunity.
Collapse
Affiliation(s)
- Xiufen Zheng
- Department of Surgery, University of Western Ontario, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Suzuki M, Zheng X, Zhang X, Zhang ZX, Ichim TE, Sun H, Nakamura Y, Inagaki A, Beduhn M, Shunnar A, Garcia B, Min WP. A novel allergen-specific therapy for allergy using CD40-silenced dendritic cells. J Allergy Clin Immunol 2010; 125:737-43, 743.e1-743.e6. [PMID: 20226305 DOI: 10.1016/j.jaci.2009.11.042] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2009] [Revised: 11/05/2009] [Accepted: 11/06/2009] [Indexed: 11/24/2022]
Abstract
BACKGROUND Induction of RNA interference with small interfering RNA (siRNA) has demonstrated therapeutic potential through the knockdown of target genes. We have previously reported that systemic administration of CD40 siRNA is capable of attenuating allergic symptoms but in an allergen-nonspecific fashion. However, siRNA-based allergen-specific therapy for allergy has not been developed. OBJECTIVE We attempted to develop a new allergen-specific therapy for allergy using CD40-silenced and allergen-pulsed dendritic cells (DCs). METHODS Bone marrow-derived DCs were silenced with CD40 siRNA and pulsed with ovalbumin (OVA). Mice had allergy after intraperitoneal sensitization with OVA and keyhole limpet hemocyanin, followed by intranasal challenge with the same allergens. The mice were treated with CD40-silenced and OVA-pulsed DCs (CD40-silenced OVA DCs) either before allergic sensitization or after establishing allergic rhinitis. RESULTS Mice receiving CD40-silenced OVA DCs either before or after the establishment of allergic rhinitis showed remarkable reductions in allergic symptoms caused by OVA challenge, as well as anti-OVA IgE levels in sera. Additionally, CD40-silenced OVA DCs suppressed eosinophil infiltration at the nasal septum, OVA-specific T-cell responses, T-cell production of IL-4 and IL-5 after stimulation with OVA, and CD4(+)CD25(-) effector T-cell responses. Furthermore, CD40-silenced OVA DCs facilitated the generation of CD4(+)CD25(+) forkhead box protein 3-positive OVA-specific regulatory T cells, which inhibit allergic responses in vivo. However, CD40-silenced OVA DCs suppressed only OVA-specific allergy but did not inhibit keyhole limpet hemocyanin-induced allergy, suggesting that CD40-silenced OVA DCs induce allergen-specific tolerance. CONCLUSIONS This study is the first to demonstrate a novel allergen-specific therapy for allergy through DC-mediated immune modulation after gene silencing of CD40.
Collapse
Affiliation(s)
- Motohiko Suzuki
- Departments of Surgery, Pathology, Microbiology, and Immunology, University of Western Ontario, Ontario, Canada.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Hilkens CMU, Isaacs JD, Thomson AW. Development of dendritic cell-based immunotherapy for autoimmunity. Int Rev Immunol 2010; 29:156-83. [PMID: 20199240 DOI: 10.3109/08830180903281193] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Dendritic cells are professional antigen-presenting cells that maintain immune tolerance to self-antigens by deleting or controlling the pathogenicity of auto-reactive T-cells. Dendritic cell-based immunotherapies show great promise for the restoration of tolerance in autoimmune disease. Dendritic cells can be modified ex vivo to induce stable tolerogenic function and be used as cellular 'vaccines' or they can be targeted in vivo with sophisticated antigen delivery systems. Tolerogenic dendritic cells induce antigen-specific T-cell tolerance in vivo and have therapeutic effects in animal models of autoimmunity. The current challenge is to bring tolerogenic dendritic cell therapy to the clinic.
Collapse
Affiliation(s)
- Catharien M U Hilkens
- Institute of Cellular Medicine, Musculoskeletal Research Group, Newcastle University, Newcastle upon Tyne, United Kingdom.
| | | | | |
Collapse
|
35
|
Suzuki JI, Ogawa M, Takayama K, Taniyama Y, Morishita R, Hirata Y, Nagai R, Isobe M. Ultrasound-Microbubble–Mediated Intercellular Adhesion Molecule-1 Small Interfering Ribonucleic Acid Transfection Attenuates Neointimal Formation After Arterial Injury in Mice. J Am Coll Cardiol 2010; 55:904-13. [DOI: 10.1016/j.jacc.2009.09.054] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Revised: 09/03/2009] [Accepted: 09/15/2009] [Indexed: 01/19/2023]
|
36
|
Zheng X, Suzuki M, Zhang X, Ichim TE, Zhu F, Ling H, Shunnar A, Wang MH, Garcia B, Inman RD, Min WP. RNAi-mediated CD40-CD154 interruption promotes tolerance in autoimmune arthritis. Arthritis Res Ther 2010; 12:R13. [PMID: 20102615 PMCID: PMC2875641 DOI: 10.1186/ar2914] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Revised: 12/08/2009] [Accepted: 01/26/2010] [Indexed: 01/25/2023] Open
Abstract
INTRODUCTION We have previously demonstrated that ex vivo inhibition of costimulatory molecules on antigen-pulsed dendritic cells (DCs) can be useful for induction of antigen-specific immune deviation and suppression of autoimmune arthritis in the collagen induced arthritis (CIA) model. The current study evaluated a practical method of immune modulation through temporary systemic inhibition of the costimulatory molecule CD40. METHODS Mice with collagen II (CII)-induced arthritis (CIA) were administered siRNA targeting the CD40 molecule. Therapeutic effects were evaluated by clinical symptoms, histopathology, Ag-specific T cell and B cell immune responses. RESULTS Systemic administration of CD40-targeting siRNA can inhibit antigen-specific T cell response to collagen II, as well as prevent pathogenesis of disease in both a pre- and post-immunization manner in the CIA model. Disease amelioration was associated with suppression of Th1 cytokines, attenuation of antibody production, and upregulation of T regulatory cells. CONCLUSIONS These studies support the feasibility of transient gene silencing at a systemic level as a mechanism of resetting autoreactive immunity.
Collapse
Affiliation(s)
- Xiufen Zheng
- Departments of Surgery, Pathology, Microbiology and Immunology, University of Western Ontario, 1393 Western Road, London, Ontario, N6G 1G9, Canada.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Ichim TE, Harman RJ, Min WP, Minev B, Solano F, Rodriguez JP, Alexandrescu DT, De Necochea-Campion R, Hu X, Marleau AM, Riordan NH. Autologous stromal vascular fraction cells: A tool for facilitating tolerance in rheumatic disease. Cell Immunol 2010; 264:7-17. [DOI: 10.1016/j.cellimm.2010.04.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Revised: 04/05/2010] [Accepted: 04/06/2010] [Indexed: 12/29/2022]
|
38
|
Zheng X, Vladau C, Shunner A, Min WP. siRNA specific delivery system for targeting dendritic cells. Methods Mol Biol 2010; 623:173-188. [PMID: 20217551 DOI: 10.1007/978-1-60761-588-0_11] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
siRNA therapy offers immense potential for clinical application. Under physiological conditions, however, siRNA was demonstrated to have a short half-life. Additionally, it may also cause ubiquitous gene silencing as it does not possess a tissue-specific homing mechanism. Thus, the rate-limiting step in the emergence of siRNA as a potential therapeutic agent is the current lack of a safe and tissue- or cell-specific in vivo delivery system. Herein, we propose a novel, cell-specific method for the in vivo delivery of siRNA to dendritic cells (DCs) with the purpose of inducing immune modulation. CD40 siRNA was incorporated within the interior of 86 nm liposomes, which were decorated with surface-bound mAb NLDC-145 as a targeting mechanism. The siRNA encapsulation efficiency was determined to be approximately 7%. CD40 siRNA immunoliposomes (CD40 siILs) were able to specifically bind to DCs and silence CD40 expression in vitro. Furthermore, in vitro CD40-silenced DCs significantly inhibited the proliferation of alloreactive T cells in an MLR. Upon in vivo administration, siIL-encapsulated, Cy3-labeled siRNA exhibited moderate uptake by the liver at an early time point following administration with greater accumulation in the spleen at a later time point. In contrast, naked siRNA primarily accumulated in the kidney immediately after administration and circulated out in a short time period. To address in vivo gene silencing and immune modulation, mice were simultaneously immunized with KLH and subcutaneously injected with DC-specific CD40 siILs, siILs containing negative control siRNA, naked CD40 siRNA, or PBS. A second injection of CD40 siILs, or control treatments, followed 24 h later. Flow cytometry, reverse transcriptase PCR, and quantitative real-time PCR analysis of CD11c(+) DCs from mice treated with CD40 siILs demonstrated reduced expression of CD40, in comparison with control groups. CD11c(-) cells were also analyzed by flow cytometry, but no differences were observed between treatment groups. Furthermore, CD40 siIL-treated mice were found to have an increased proportion of Treg cells (CD4(+)CD25(+) FoxP3(+)), and DCs cells from these mice were able to inhibit T cell proliferation in an antigen-specific recall response. In summary, CD40 siILs were shown to specifically target and deliver CD40 siRNA to DCs, significantly reducing CD40 expression and resulting in DC-mediated immune modulation as well as generation of Treg cells. These findings highlight the therapeutic potential for siRNA-based and DC-mediated immunotherapy in the clinic. To the best of our knowledge, this is the first study to use siILs for targeted delivery of siRNA to DCs and for immune modulation.
Collapse
Affiliation(s)
- Xiufen Zheng
- Department of Surgery, University of Western Ontario, London, ON, Canada
| | | | | | | |
Collapse
|
39
|
Nemunaitis J, Roth J. Gene-Based Therapies for Lung Cancer. Lung Cancer 2010. [DOI: 10.1007/978-1-60761-524-8_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
40
|
Abstract
Dendritic cells (DCs) comprise a family of professional antigen-presenting cells responsible for the induction of primary immune responses. DCs are also important for the induction of immunological tolerance. Recent research has revealed that DC maturation is associated with activation of the NF-kappaB pathway. RelB, one of the five families of Rel proteins involved in the NF-kappaB pathway, plays a critical role in coordinating the terminal stages of DC maturation and has the ability to induce optimal Th1 T cell responses. DCs generated from mouse bone marrow can be silenced using siRNA specific for the target gene. Silencing RelB in DCs will result in the generation of immunoregulatory dendritic cells that inhibit allogenic T cell responses. The KLH-specific T cell response should also be inhibited after the RelB siRNA treatment. Furthermore, silencing the RelB gene in DCs can generate regulatory T cells. Administering donor-derived RelB-silencing DCs can prevent allograft rejection in murine heart transplantation.
Collapse
Affiliation(s)
- Xusheng Zhang
- Department of Surgery, University of Western Ontario, London, ON, Canada
| | | | | |
Collapse
|
41
|
Okamoto M, Matsuda H, Joetham A, Lucas JJ, Domenico J, Yasutomo K, Takeda K, Gelfand EW. Jagged1 on Dendritic Cells and Notch on CD4+ T Cells Initiate Lung Allergic Responsiveness by Inducing IL-4 Production. THE JOURNAL OF IMMUNOLOGY 2009; 183:2995-3003. [DOI: 10.4049/jimmunol.0900692] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
42
|
Herringson TP, Altin JG. Convenient targeting of stealth siRNA-lipoplexes to cells with chelator lipid-anchored molecules. J Control Release 2009; 139:229-38. [PMID: 19595724 DOI: 10.1016/j.jconrel.2009.06.034] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 05/18/2009] [Accepted: 06/30/2009] [Indexed: 01/03/2023]
Abstract
A major obstacle for the use of siRNAs as novel therapeutics is the requirement for functional delivery to specific cells in vivo. siRNA delivery by cationic agents is generally non-specific and a convenient targeting strategy has been lacking. This work explored the potential for using the chelator lipid 3(nitrilotriacetic acid)-ditetradecylamine (NTA(3)-DTDA) with neutral stealth liposomes to target siRNA to cells. A novel method for incorporating siRNAs into lipoplexes was developed which utilised helper lipids and the ionisable lipid 1,2-dioleoyl-3-dimethylammonium-propane (DODAP). This approach results in an efficient (>50%) incorporation of siRNA into lipoplexes, which when incorporated with Ni-NTA(3)-DTDA and engrafted with a His-tagged form of murine CD4 can target siRNA to murine A20 B cells, in vitro. Also, siRNA-lipoplexes engrafted with His-tagged peptides that target receptors on HEK-293 cells, or the receptor for tumour necrosis factor alpha expressed on the murine dendritic cell line DC2.4, could target siRNA and silence the expression of enhanced green fluorescence protein (EGFP). siRNA-lipoplexes produced by this method are approximately 240 nm dia, exhibit low zeta-potential (-1 mV), and target cells in serum-containing media. The results show that NTA(3)-DTDA can be used to target siRNA-lipoplexes to cells, and could provide a convenient approach for targeting siRNA to cells in vivo for therapeutic applications.
Collapse
Affiliation(s)
- Thomas P Herringson
- Biochemistry and Molecular Biology, Research School of Biology, ANU College of Medicine, Biology and Environment, The Australian National University, Canberra, ACT, 0200, Australia
| | | |
Collapse
|
43
|
Ghafoori P, Yoshimura T, Turpie B, Masli S. Increased IkappaB alpha expression is essential for the tolerogenic property of TGF-beta-exposed APCs. FASEB J 2009; 23:2226-34. [PMID: 19237504 PMCID: PMC2704595 DOI: 10.1096/fj.08-124545] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Accepted: 01/29/2009] [Indexed: 11/11/2022]
Abstract
IkappaB alpha is an inhibitor of the transcriptional factor NF-kappaB, and it is an essential component of the signaling pathways that lead to expression of inflammatory molecules. These include cytokines and costimulatory molecules associated with antigen presentation in an inflammatory immune response. In this study, we report that antigen-presenting cells exposed to TGF-beta induce peripheral tolerance by increasing IkappaB alpha expression. Exposure of antigen presenting cells (APCs) to TGF-beta is known to impair their ability to secrete IL-12, and such impairment correlated with reduced NF-kappaB activity as indicated by significantly reduced nuclear levels of p50, an essential subunit of NF-kappaB for IL-12 transcription. Blockade of increased nuclear IkappaB alpha in APCs by expression of small interfering RNA molecules (siRNAs) targeting IkappaB alpha transcripts prevented IL-12 impairment and the decline in nuclear p50 levels. Furthermore, such IkappaB alpha blockade also interfered with the tolerogenic property of TGF-beta-exposed APCs. However, increased expression of IkappaB alpha in APCs, independent of TGF-beta exposure, reduced nuclear p50 levels and permitted tolerance induction by APCs. Thus, our findings attribute a direct and significant role to IkappaB alpha in the tolerogenic potential of APCs. Increased IkappaB alpha expression in APCs may therefore offer a therapeutic approach to achieve antigen-specific immunomodulation.
Collapse
Affiliation(s)
- Paiman Ghafoori
- Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | | | | | | |
Collapse
|
44
|
Boudier A, Aubert-Pouëssel A, Gérardin C, Devoisselle JM, Bégu S, Louis-Plence P, Quentin J, Jorgensen C. Tripartite siRNA micelles as controlled delivery systems for primary dendritic cells. Drug Dev Ind Pharm 2009; 35:950-8. [DOI: 10.1080/03639040802716653] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
45
|
Both treated and untreated tumors are eliminated by short hairpin RNA-based induction of target-specific immune responses. Proc Natl Acad Sci U S A 2009; 106:8314-9. [PMID: 19416823 DOI: 10.1073/pnas.0812085106] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
RNA interference (RNAi) for cancer treatment relies on the ability to directly kill cancer cells via down-regulation of target genes, but issues of delivery and efficacy have limited clinical adoption. Furthermore, current studies using immune-deficient animal models disregard potential interactions with the adaptive immune system. It has previously been observed that certain viral antigens appear to be more rapidly presented to the immune system than normal proteins due to the production of defective ribosomal products by the virus. Given that RNAi could potentially result in the generation of truncated mRNAs, we wondered whether a similar mechanism of immune presentation of a target gene was possible. Here we show that RNAi-cleaved mRNAs can be translated into incomplete protein, and if cleavage was downstream of cytotoxic T cell epitopes, resulted in increased presentation of target protein and the generation of a tumor-protective immune response. We show that mice inoculated with tumor cells treated with such short hairpin RNAs (shRNAs) were protected from subsequent challenge with untreated tumors. However, protection was only found if shRNAs were targeted downstream of the dominant cytotoxic T cell (CTL) epitope. Our work suggests that RNAi can alter immunity to targets and shows that not all tumor cells require direct RNAi exposure for treatment to be effective in vivo, pointing the way to a new class of RNAi-based therapy.
Collapse
|
46
|
Zhang L, Procuik M, Fang T, Kung SKP. Functional analysis of the quantitative expression of a costimulatory molecule on dendritic cells using lentiviral vector-mediated RNA interference. J Immunol Methods 2009; 344:87-97. [PMID: 19303417 DOI: 10.1016/j.jim.2009.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2008] [Revised: 03/05/2009] [Accepted: 03/10/2009] [Indexed: 12/13/2022]
Abstract
The increasing number of co-stimulatory molecules identified on dendritic cells (DC) to date highlights the complex regulation of co-stimulatory signals in T cell activation. We previously established a single lentiviral vector system to stably express short hairpin RNA (shRNA) to induce RNA interference (RNAi) in cell lines and primary T cells. We reasoned that the choice of shRNA target sequences in the lentiviral vector system would also allow us to regulate different levels of surface expression of a co-stimulatory molecule stably and reproducibly. In this study, we first demonstrated that lentiviral vectors delivered RNA interference in DC without functional impairments. We used CD40 as a target co-stimulatory molecule to demonstrate the feasibility of using lentiviral vectors in delivering different shRNA target sequences to genetically modify DC that expressed different levels of CD40. We provided functional data to further demonstrate that quantitative expression of CD40 on LPS-stimulated DC have different functional outcomes on Ag-specific T cell responses in vitro. Collectively, we developed a simple system that will allow us to examine functional significance(s) of the quantitative and/or qualitative expression of a single or multiple co-stimulatory molecule(s) on DC.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Immunology, University of Manitoba, Room 417, Apotex Center, 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E0T5
| | | | | | | |
Collapse
|
47
|
Suzuki M, Zheng X, Zhang X, Ichim TE, Sun H, Kubo N, Beduhn M, Shunnar A, Garcia B, Min WP. Inhibition of allergic responses by CD40 gene silencing. Allergy 2009; 64:387-97. [PMID: 19175598 DOI: 10.1111/j.1398-9995.2008.01839.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Gene silencing using small interfering RNA (siRNA) is a potent method of specifically knocking down molecular targets. Small interfering RNA is therapeutically promising, however, treatment of allergic diseases with siRNA has not been explored in vivo. The aim of this study was to evaluate therapeutic effects of CD40 siRNA on inhibition of allergic responses. METHODS Mice sensitized with ovalbumin (OVA) and alum were treated with CD40 siRNA, scrambled siRNA, or phosphate buffer saline (PBS) alone, and then challenged intranasally with OVA. RESULTS A significant reduction in nasal allergic symptoms was observed in the CD40 siRNA treated OVA-allergic mice compared to the controls of scrambled siRNA and PBS alone, which is correlated with the decrease of local eosinophil accumulation. CD40 siRNA treatment knocked down CD40 expression on dendritic cells (DCs) in vivo and impaired their antigen presenting function. Treatment with CD40 siRNA resulted in inhibition of OVA-specific T cell response and decrease of interleukin-4 (IL-4), IL-5, and interferon-gamma production from T cells stimulated with OVA. Administration of CD40 siRNA also suppressed CD40 expression on B cells, resulting in down-regulation of OVA-specific immunoglobulin E (IgE), IgG1, and IgG2a levels. Additionally, increased regulatory T cells were observed in the CD40 siRNA treated mice. CONCLUSIONS The present study demonstrates a novel therapeutic use for siRNA in allergy. CD40 siRNA attenuated allergy through inhibition of DC and B cell functions and generation of regulatory T (Treg) cells.
Collapse
Affiliation(s)
- M Suzuki
- Department of Surgery, University of Western Ontario, London, ON, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
A novel in vivo siRNA delivery system specifically targeting dendritic cells and silencing CD40 genes for immunomodulation. Blood 2009; 113:2646-54. [PMID: 19164600 DOI: 10.1182/blood-2008-04-151191] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Translation of small interfering RNA (siRNA)-based approaches into practical therapeutics is limited because of lack of an effective and cell-specific delivery system. Herein, we present a new method of selectively delivering siRNA to dendritic cells (DCs) in vivo using CD40 siRNA-containing immunoliposomes (siILs) that were decorated with DC-specific DEC-205 mAb. Administration of CD40 siILs resulted in DC-specific cell targeting in vitro and in vivo. On treatment with CD40 siILs, the expression of CD40 in DCs, as well allostimulatory activity was inhibited. In vivo administration resulted in selective siRNA uptake into immune organs and functional immune modulation as assessed using a model antigen. In conclusion, this is the first demonstration of DC-specific siRNA delivery and gene silencing in vivo, which highlights the potential of DC-mediated immune modulation and the feasibility of siRNA-based clinical therapy.
Collapse
|
49
|
Karimi MH, Ebadi P, Pourfathollah AA, Soheili ZS, Samiee S, Ataee Z, Tabei SZ, Moazzeni SM. Immune modulation through RNA interference-mediated silencing of CD40 in dendritic cells. Cell Immunol 2009; 259:74-81. [DOI: 10.1016/j.cellimm.2009.05.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2009] [Revised: 05/12/2009] [Accepted: 05/22/2009] [Indexed: 11/30/2022]
|
50
|
Kohno K, Miyake M, Sano O, Tanaka-Kataoka M, Yamamoto S, Koya-Miyata S, Arai N, Fujii M, Watanabe H, Ushio S, Iwaki K, Fukuda S. Anti-inflammatory and immunomodulatory properties of 2-amino-3H-phenoxazin-3-one. Biol Pharm Bull 2008; 31:1938-45. [PMID: 18827359 DOI: 10.1248/bpb.31.1938] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Accumulating evidence suggests that nitric oxide (NO) and prostaglandin E(2) (PGE(2)) are involved in the pathogenesis of various chronic inflammatory diseases and cancer. During the course of a screening program to identify natural anti-inflammatory substances, we isolated the compound 2-amino-3H-phenoxazin-3-one (APO) from an extract of the edible brown mushroom Agaricus bisporus IMBACH. APO inhibited NO production by mouse peritoneal macrophages in response to the pro-inflammatory stimuli lipopolysaccharide (LPS) and interferon (IFN)-gamma (LPS/IFN-gamma) at low concentrations (IC(50)=1.5 microM) through reduced inducible NO synthase protein expression. PGE(2) production by LPS/IFN-gamma-stimulated macrophages was inhibited by APO at much lower concentrations (IC(50)=0.27 microM) than those required for the inhibition of NO production. Mechanistic analysis showed that APO inhibited both cyclooxygenase (COX)-1 and COX-2 enzyme activities with almost equal selectivity. Secretion of NO and the pro-inflammatory cytokine IL-6 by IFN-gamma-activated RAW264.7 cells, a murine macrophage-like cell line, was also dose-dependently reduced by APO. Furthermore, APO increased the secretion of the anti-inflammatory cytokine IL-4 by antigen-stimulated T cells and promoted the polarization of CD4(+) Th cells toward the anti-inflammatory Th2 phenotype at equimolar concentrations that inhibited NO production. Our results suggested that APO induced polarization toward the Th2 subset, at least in part through the down-regulation of IL-12 production. Thus, APO appears to have potent anti-inflammatory and immunoregulatory properties that may provide a promising therapeutic strategy for the treatment of T cell-mediated inflammatory autoimmune diseases as well as for bacteria-induced chronic-inflammatory diseases.
Collapse
Affiliation(s)
- Keizo Kohno
- Biomedical Institute, Research Center, Hayashibara Biochemical Laboratories, Inc, 675-1 Fujisaki, Okayama 702-8006, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|