1
|
Wang YZ, Yao-Li, Liang SK, Ding LB, Feng-Li, Guan J, Wang HJ. LncPVT1 promotes cartilage degradation in diabetic OA mice by downregulating miR-146a and activating TGF-β/SMAD4 signaling. J Bone Miner Metab 2021; 39:534-546. [PMID: 33569722 DOI: 10.1007/s00774-020-01199-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 12/14/2020] [Indexed: 10/22/2022]
Abstract
INTRODUCTION To investigate the role of LncRNA PVT1 (plasmacytoma variant translocation 1) in hyperglycemia-triggered cartilage damage using the diabetic osteoarthritis (OA) mice model. MATERIALS AND METHODS Streptozotocin (STZ) was used to induce mouse diabetes. Knee OA model was induced through transection of anterior cruciate ligament (ACLT). Severity of arthritis was assessed histologically by Safranin O-Fast Green Staining using Mankin Scores. LncRNA PVT1 and miR-146a were detected by real-time polymerase chain reaction (PCR) in cartilage tissue. Moreover, the interaction among PVT1, miR-146a, and SMAD4 was examined by luciferase reporter assays. Mice were injected intra-articularly with ad-siRNA-PVT1 and ad-siRNA scramble control. Articular concentrations of TNF-α, IL-1, IL-6 and TGF-β1 were determined using enzyme-linked immunosorbent assay. Levels of type II Collagen (COL2A1), TGF-β1, p-SMAD2, SMAD2, p-SMAD3, SMAD3, SMAD4 and nuclear SMAD4 were detected by western blot analysis. RESULTS PVT1 expression was significantly increased, whereas miR-146a was markedly decreased in diabetic OA mice than in non-diabetic OA and control. Increased PVT1 expression in diabetic OA mice was significantly associated with Mankin score and reduced miR-146a as well as Collagen alpha-1(II) (COL2A1) expressions. In vivo, intra-articular injection of ad-siRNA-PVT1 efficiently increased miR-146a and COL2A1 expressions, alleviated joint inflammation, decreased the expression of pro-inflammatory mediators, and suppressed TGF-β/SMAD4 pathway in diabetic OA mice. CONCLUSIONS Our results demonstrate LncRNA PVT1 is involved in cartilage degradation in diabetic OA and correlated with disease severity. Efficiency of ad-siRNA-PVT1 in controlling joint inflammation in diabetic OA mice is associated with the suppression of the expression of miR-146a, pro-inflammatory cytokines and activation of TGF-β/SMAD4 pathway.
Collapse
Affiliation(s)
- Yan-Zhi Wang
- Department of Orthopedic Surgery, Third Hospital of Shijiazhuang, ChangAn District, , Shijiazhuang, 050011, HeBei, China
| | - Yao-Li
- Department of Orthopedic Surgery, Third Hospital of Shijiazhuang, ChangAn District, , Shijiazhuang, 050011, HeBei, China
| | - Sheng-Kai Liang
- Department of Orthopedic Surgery, Third Hospital of Shijiazhuang, ChangAn District, , Shijiazhuang, 050011, HeBei, China
| | - Luo-Bin Ding
- Department of Orthopedic Surgery, Third Hospital of Shijiazhuang, ChangAn District, , Shijiazhuang, 050011, HeBei, China
| | - Feng-Li
- Department of Orthopedic Surgery, Third Hospital of Shijiazhuang, ChangAn District, , Shijiazhuang, 050011, HeBei, China
| | - Jian Guan
- Department of Orthopedic Surgery, Third Hospital of Shijiazhuang, ChangAn District, , Shijiazhuang, 050011, HeBei, China.
| | - Hua-Jun Wang
- Department of Orthopedic Surgery and Sports Medicine Center, The First Affiliated Hospital of Jinan University, Road West No. 613, Huang-Pu, Guangzhou, 510632, GuangDong, China.
| |
Collapse
|
2
|
Leng Q, Chen L, Lv Y. RNA-based scaffolds for bone regeneration: application and mechanisms of mRNA, miRNA and siRNA. Am J Cancer Res 2020; 10:3190-3205. [PMID: 32194862 PMCID: PMC7053199 DOI: 10.7150/thno.42640] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 01/16/2020] [Indexed: 02/07/2023] Open
Abstract
Globally, more than 1.5 million patients undergo bone graft surgeries annually, and the development of biomaterial scaffolds that mimic natural bone for bone grafting remains a tremendous challenge. In recent decades, due to the improved understanding of the mechanisms of bone remodeling and the rapid development of gene therapy, RNA (including messenger RNA (mRNA), microRNA (miRNA), and short interfering RNA (siRNA)) has attracted increased attention as a new tool for bone tissue engineering due to its unique nature and great potential to cure bone defects. Different types of RNA play roles via a variety of mechanisms in bone-related cells in vivo as well as after synthesis in vitro. In addition, RNAs are delivered to injured sites by loading into scaffolds or systemic administration after combination with vectors for bone tissue engineering. However, the challenge of effectively and stably delivering RNA into local tissue remains to be solved. This review describes the mechanisms of the three types of RNAs and the application of the relevant types of RNA delivery vectors and scaffolds in bone regeneration. The improvements in their development are also discussed.
Collapse
|
3
|
Paganin-Gioanni A, Rols MP, Teissié J, Golzio M. Cyclin B1 knockdown mediated by clinically approved pulsed electric fields siRNA delivery induces tumor regression in murine melanoma. Int J Pharm 2020; 573:118732. [DOI: 10.1016/j.ijpharm.2019.118732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 09/18/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023]
|
4
|
Adenovirus-Mediated Small Interfering RNA Targeting TAK1 Ameliorates Joint Inflammation with Collagen-Induced Arthritis in Mice. Inflammation 2018; 40:894-903. [PMID: 28220341 DOI: 10.1007/s10753-017-0534-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Transforming growth factor β-activated kinase-1 (TAK1) is a key upstream kinase in cell signaling during inflammation, which regulates the expression of inflammatory mediators. Small interfering RNA (siRNA) against TAK1 offers promise as a potential therapeutic strategy in immune-mediated inflammatory disorder including rheumatoid arthritis. Here, we are to evaluate the therapeutic effects of intra-articular administration of adenoviral-mediated siRNA against TAK1 (ad-siRNA-TAK1) on collagen-induced arthritis (CIA) in mice. Ad-siRNA-TAK1 was constructed. The murine RAW 264.7 macrophages were infected with ad-siRNA-TAK1, and the silencing specificity of TAK1 was assessed by quantitative polymerase chain reaction (PCR) and western blot. DBA/1 mice were injected intra-articularly with ad-siRNA-TAK1. Development and severity of arthritis was assessed histologically. Synovial inflammation and bone destruction were determined by hematoxylin and eosin (HE) staining. Articular and serum concentrations of tumor necrosis factor-α, interleukin-1, and interleukin-6 were determined using enzyme-linked immunosorbent assay. Levels of phosphorylated p38, c-Jun N-terminal kinase (JNK), and extracellular-signal-regulated kinase (ERK) were detected by western blot. In vitro, ad--siRNA-TAK1 efficiently inhibited the expression of TAK1 at both mRNA and protein levels. In vivo, intra-articular injection of ad-siRNA-TAK1 efficiently alleviated joint inflammation, decreased the expression of pro-inflammatory mediators, and suppressed JNK pathways. Our results demonstrate the efficiency of ad--siRNA-TAK1 in controlling joint inflammation of CIA, which is associated with the suppression of the expression of pro-inflammatory cytokines and JNK activation.
Collapse
|
5
|
Banda NK, Acharya S, Scheinman RI, Mehta G, Coulombe M, Takahashi M, Sekine H, Thiel S, Fujita T, Holers VM. Mannan-Binding Lectin-Associated Serine Protease 1/3 Cleavage of Pro-Factor D into Factor D In Vivo and Attenuation of Collagen Antibody-Induced Arthritis through Their Targeted Inhibition by RNA Interference-Mediated Gene Silencing. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:3680-3694. [PMID: 27707997 PMCID: PMC5113144 DOI: 10.4049/jimmunol.1600719] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 09/07/2016] [Indexed: 12/16/2022]
Abstract
The complement system is proposed to play an important role in the pathogenesis of rheumatoid arthritis (RA). The complement system mannan-binding lectin-associated serine proteases (MASP)-1/3 cleave pro-factor D (proDf; inactive) into Df (active), but it is unknown where this cleavage occurs and whether inhibition of MASP-1/3 is a relevant therapeutic strategy for RA. In the present study, we show that the cleavage of proDf into Df by MASP-1/3 can occur in the circulation and that inhibition of MASP-1/3 by gene silencing is sufficient to ameliorate collagen Ab-induced arthritis in mice. Specifically, to examine the cleavage of proDf into Df, MASP-1/3-producing Df-/- liver tissue (donor) was transplanted under the kidney capsule of MASP-1/3-/- (recipient) mice. Five weeks after the liver transplantation, cleaved Df was present in the circulation of MASP-1/3-/- mice. To determine the individual effects of MASP-1/3 and Df gene silencing on collagen Ab-induced arthritis, mice were injected with scrambled, MASP-1/3-targeted, or Df-targeted small interfering RNAs (siRNAs). The mRNA levels for MASP-1 and -3 decreased in the liver to 62 and 58%, respectively, in mice injected with MASP-1/3 siRNAs, and Df mRNA decreased to 53% in the adipose tissue of mice injected with Df siRNAs; additionally, circulating MASP-1/3 and Df protein levels were decreased. In mice injected with both siRNAs the clinical disease activity, histopathologic injury scores, C3 deposition, and synovial macrophage/neutrophil infiltration were significantly decreased. Thus, MASP-1/3 represent a new therapeutic target for the treatment of RA, likely through both direct effects on the lectin pathway and indirectly through the alternative pathway.
Collapse
Affiliation(s)
- Nirmal K Banda
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045;
| | - Sumitra Acharya
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Robert I Scheinman
- School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Gaurav Mehta
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Marilyne Coulombe
- Colorado Center for Transplantation Care, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Minoru Takahashi
- Department of Immunology, Fukushima Medical University, Fukushima 960-1295, Japan; and
| | - Hideharu Sekine
- Department of Immunology, Fukushima Medical University, Fukushima 960-1295, Japan; and
| | - Steffen Thiel
- Department of Biomedicine, University of Aarhus, 8000 Aarhus, Denmark
| | - Teizo Fujita
- Department of Immunology, Fukushima Medical University, Fukushima 960-1295, Japan; and
| | - V Michael Holers
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
6
|
Targeted electro-delivery of oligonucleotides for RNA interference: siRNA and antimiR. Adv Drug Deliv Rev 2015; 81:161-8. [PMID: 24819217 DOI: 10.1016/j.addr.2014.05.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 04/18/2014] [Accepted: 05/01/2014] [Indexed: 12/11/2022]
Abstract
For more than a decade, the understanding of RNA interference (RNAi) has been a growing field of interest. Micro-RNAs (miRNAs) are small regulatory RNAs that play an important role in disease development and progression and therefore represent a potential new class of therapeutic targets. However, delivery of RNAi-based oligonucleotides is one of the most challenging hurdles to RNAi-based drug development. Electropermeabilization (EP) is recognized as a successful non-viral method to transfer nucleic acids into living cells both in vitro and in vivo. EP is the direct application of electric pulses to cells or tissues that transiently permeabilize plasma membranes, allowing the efficient delivery of exogenous molecules. The present review focused on the mechanism of RNAi-based oligonucleotides electrotransfer, from cellular uptake to intracellular distribution. Biophysical theories on oligonucleotide electrotransfer will be also presented. The advantages and few drawbacks of EP-mediated delivery will also be discussed.
Collapse
|
7
|
Duan H, Yang P, Fang F, Ding S, Xiao W. CCR5 small interfering RNA ameliorated joint inflammation in rats with adjuvant-induced arthritis. Immunol Lett 2014; 162:258-63. [DOI: 10.1016/j.imlet.2014.09.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 09/26/2014] [Accepted: 09/26/2014] [Indexed: 10/24/2022]
|
8
|
Akagi R, Sasho T, Saito M, Endo J, Yamaguchi S, Muramatsu Y, Mukoyama S, Akatsu Y, Katsuragi J, Fukawa T, Takahashi K. Effective knock down of matrix metalloproteinase-13 by an intra-articular injection of small interfering RNA (siRNA) in a murine surgically-induced osteoarthritis model. J Orthop Res 2014; 32:1175-80. [PMID: 24848439 DOI: 10.1002/jor.22654] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 05/02/2014] [Indexed: 02/04/2023]
Abstract
This study investigated the effect of MMP-13 gene knock down on cartilage degradation by injecting small interfering RNA (siRNA) into knee joints in a mouse model of osteoarthritis (OA). OA was induced in male C57BL/6 mice by destabilization of medial meniscus (DMM) surgery. Change of Mmp13 expression over time was determined by qPCR analysis from 3 days to 6 weeks after surgery. Mmp13 and control chemically modified siRNA were injected into the knee joint 1 week after surgery and expression levels were assessed in synovium by qPCR 48 h later. Cartilage degradation was histologically assessed 8 weeks after DMM surgery according to OARSI recommendations. Mmp13 expression levels were elevated 1 week after surgery and peaked at 77 fold at 2 weeks compared to expression at 3 days. A 55% decrease of Mmp13 levels in cartilage was observed 48 h after injection of Mmp13 siRNA (p = 0.05). Significant reduction in the histological score at 8 weeks after surgery was observed in the Mmp13 siRNA-treated group compared to the control siRNA group (p < 0.001). Intra-articular injection of Mmp13 siRNA at the early phase of OA development resulted in effective knock down of Mmp13 expression and delay in cartilage degradation in vivo.
Collapse
Affiliation(s)
- Ryuichiro Akagi
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana Chuo-ku, Chiba, Chiba, 260-8670, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Inoue H, Arai Y, Kishida T, Shin-Ya M, Terauchi R, Nakagawa S, Saito M, Tsuchida S, Inoue A, Shirai T, Fujiwara H, Mazda O, Kubo T. Sonoporation-mediated transduction of siRNA ameliorated experimental arthritis using 3 MHz pulsed ultrasound. ULTRASONICS 2014; 54:874-881. [PMID: 24291002 DOI: 10.1016/j.ultras.2013.10.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 10/28/2013] [Accepted: 10/28/2013] [Indexed: 06/02/2023]
Abstract
The goal of this feasibility study was to examine whether sonoporation assisted transduction of siRNA could be used to ameliorate arthritis locally. If successful, such approach could provide an alternative treatment for the patients that have or gradually develop adverse response to chemical drugs. Tumor necrosis factor alpha (TNF-α) produced by synovial fibroblasts has an important role in the pathology of rheumatoid arthritis, inducing inflammation and bone destruction. In this study, we injected a mixture of microbubbles and siRNA targeting TNF-α (siTNF) into the articular joints of rats, and transduced siTNF into synovial tissue by exposure to a collimated ultrasound beam, applied through a probe 6mm in diameter with an input frequency of 3.0 MHz, an output intensity of 2.0 W/cm(2) (spatial average temporary peak; SATP), a pulse duty ratio of 50%, and a duration of 1 min. Sonoporation increased skin temperature from 26.8 °C to 27.3 °C, but there were no adverse effect such as burns. The mean level of TNF-α expression in siTNF-treated knee joints was 55% of those in controls. Delivery of siTNF into the knee joints every 3 days (i.e., 7, 10, 13, and 16 days after immunization) by in vivo sonoporation significantly reduced paw swelling on days 20-23 after immunization. Radiographic scores in the siTNF group were 56% of those in the CIA group and 61% of those in the siNeg group. Histological examination showed that the number of TNF-α positive cells was significantly lower in areas of pannus invasion into the ankle joints of siTNF- than of siNeg-treated rats. These results indicate that transduction of siTNF into articular synovium using sonoporation may be an effective local therapy for arthritis.
Collapse
Affiliation(s)
- Hiroaki Inoue
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yuji Arai
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan.
| | - Tsunao Kishida
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Masaharu Shin-Ya
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Ryu Terauchi
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Shuji Nakagawa
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Masazumi Saito
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Shinji Tsuchida
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Atsuo Inoue
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Toshiharu Shirai
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, 13-1 Takaramachi, Kanazawa 920-8641, Japan
| | - Hiroyoshi Fujiwara
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Osam Mazda
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Toshikazu Kubo
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
10
|
siRNA delivery via electropulsation: a review of the basic processes. Methods Mol Biol 2014; 1121:81-98. [PMID: 24510814 DOI: 10.1007/978-1-4614-9632-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2023]
Abstract
Due to their capacity for inducing strong and sequence specific gene silencing in cells, small interfering RNAs (siRNAs) are now recognized not only as powerful experimental tools for basic research in Molecular biology but with promising potentials in therapeutic development. Delivery is a bottleneck in many studies. There is a common opinion that full potential of siRNA as therapeutic agent will not be attained until better methodologies for its targeted intracellular delivery to cells and tissues are developed. Electropulsation (EP) is one of the physical methods successfully used to transfer siRNA into living cells in vitro and in vivo. This review will describe how siRNA electrotransfer obeys characterized biophysical processes (cell-size-dependent electropermeabilization, electrophoretic drag) with a strong control of a low loss of viability. Protocols can be easily adjusted by a proper setting of the electrical parameters and pulsing buffers. EP can be easily directly applied on animals. Preclinical studies showed that electropermeabilization brings a direct cytoplasmic distribution of siRNA and an efficient silencing of the targeted protein expression. EP appears as a promising tool for clinical applications of gene silencing. A panel of successful trials will be given.
Collapse
|
11
|
Inoue A, Takahashi KA, Mazda O, Arai Y, Saito M, Kishida T, Shin-Ya M, Morihara T, Tonomura H, Sakao K, Imanishi J, Kubo T. Comparison of anti-rheumatic effects of local RNAi-based therapy in collagen induced arthritis rats using various cytokine genes as molecular targets. Mod Rheumatol 2014. [DOI: 10.3109/s10165-008-0131-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Atsuo Inoue
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine,
465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
- Department of Orthopaedics, Kyoto First Red Cross Hospital,
Kyoto 605-0981, Japan
| | - Kenji A. Takahashi
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine,
465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Osam Mazda
- Department of Microbiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine,
465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yuji Arai
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine,
465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Masazumi Saito
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine,
465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Tsunao Kishida
- Department of Microbiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine,
465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
- Louis Pasteur Center for Medical Research,
Kyoto 606-8225, Japan
| | - Masaharu Shin-Ya
- Department of Microbiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine,
465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Toru Morihara
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine,
465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Hitoshi Tonomura
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine,
465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Kei Sakao
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine,
465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Jiro Imanishi
- Department of Microbiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine,
465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Toshikazu Kubo
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine,
465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
12
|
Lv F, Song LJ, Wang XH, Qiu F, Li XF. The role of Act1, a NF-κB-activating protein, in IL-6 and IL-8 levels induced by IL-17 stimulation in SW982 cells. PHARMACEUTICAL BIOLOGY 2013; 51:1444-1450. [PMID: 23862741 DOI: 10.3109/13880209.2013.798668] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
CONTEXT Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic inflammation in the synovial membrane of affected joints. It has been shown that several kinds of cytokine were increased in synovial fluid, while the underlying mechanism remains poorly understood. OBJECTIVES NF-κB activator 1 (Act1) is a recently identified protein binding to the IκB kinase complex. Our study aimed to investigate the expression of Act1 induced by cytokine IL-17 stimulation in SW982 cells. MATERIALS AND METHODS The human synovial sarcoma cell line SW982 and primary cultured RA fibroblast-like synovial cells were used. RT-PCR and Western blot assays were selected to investigate the genetic and protein expression of Act1. Additionally, four independent Act1 small interfering RNA (siRNA) oligonucleotides were designed and obtained according to the GenBank cDNA, the sequence of Act1 (Traf3ip2). Finally, enzyme-linked immunosorbent assay (ELISA) double antibody sandwich was used to assay supernatant IL-6 and IL-8 concentrations. RESULTS The Act1 mRNA expression level increased significantly after stimulation with IL-17 (5-100 ng/ml) in SW982 cells. Additionally, the level of Act1 mRNA expression correlated positively with the concentration of IL-17 (p < 0.01). IL-17 induced IL-6 and IL-8 in SW982 cells was in a concentration- and time-dependent way. Furthermore, ELISA assay revealed that IL-17 (20 ng/ml) significantly increased IL-6 (1927.4 ± 288.77 versus 786.5 ± 172.42 ng/ml, p < 0.01) and IL-8 levels (984.8 ± 95.09 ng/ml versus 307.1 ± 90.83 ng/ml, p < 0.01) compared with control group after stimulation for 24 h. However, transfection of Traf3ip2 siRNA markedly decreased IL-6 (995.9 ± 115.30 ng/ml versus 1816.1 ± 273.27 ng/ml, p < 0.01) and IL-8 levels (575.6 ± 65.96 ng/ml versus 929.4 ± 124.39 ng/ml, p < 0.01) compared to transfection negative control. These findings suggested that IL-6 and IL-8 level induced by IL-17 in SW982 cells could be reversed by down-regulation of Act1 expression level with Traf3ip2 siRNA. CONCLUSION Our results suggested that Act1 might play a key role in the pathophysiology and the treatment of RA.
Collapse
Affiliation(s)
- Fang Lv
- Department of Rheumatology, Qilu Hospital of Shandong University , Jinan , China
| | | | | | | | | |
Collapse
|
13
|
Chabot S, Pelofy S, Teissié J, Golzio M. Delivery of RNAi-Based Oligonucleotides by Electropermeabilization. Pharmaceuticals (Basel) 2013; 6:510-21. [PMID: 24276121 PMCID: PMC3816695 DOI: 10.3390/ph6040510] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 03/19/2013] [Accepted: 03/27/2013] [Indexed: 11/16/2022] Open
Abstract
For more than a decade, understanding of RNA interference (RNAi) has been a growing field of interest. The potent gene silencing ability that small oligonucleotides have offers new perspectives for cancer therapeutics. One of the present limits is that many biological barriers exist for their efficient delivery into target cells or tissues. Electropermeabilization (EP) is one of the physical methods successfully used to transfer small oligonucleotides into cells or tissues. EP consists in the direct application of calibrated electric pulses to cells or tissues that transiently permeabilize the plasma membranes, allowing efficient in vitro and in vivo cytoplasmic delivery of exogenous molecules. The present review reports on the type of therapeutic RNAi-based oligonucleotides that can be electrotransferred, the mechanism(s) of their electrotransfer and the technical settings for pre-clinical purposes.
Collapse
Affiliation(s)
- Sophie Chabot
- Centre National de la Recherche Scientifique (CNRS), Institut de Pharmacologie et de Biologie Structurale (IPBS) BP 64182, 205 route de Narbonne, Toulouse F-31077, France; E-Mails: (S.C.); (S.P.); (J.T.)
- Université Paul Sabatier de Toulouse, IPBS, Toulouse F-31077, France
| | - Sandrine Pelofy
- Centre National de la Recherche Scientifique (CNRS), Institut de Pharmacologie et de Biologie Structurale (IPBS) BP 64182, 205 route de Narbonne, Toulouse F-31077, France; E-Mails: (S.C.); (S.P.); (J.T.)
- Université Paul Sabatier de Toulouse, IPBS, Toulouse F-31077, France
| | - Justin Teissié
- Centre National de la Recherche Scientifique (CNRS), Institut de Pharmacologie et de Biologie Structurale (IPBS) BP 64182, 205 route de Narbonne, Toulouse F-31077, France; E-Mails: (S.C.); (S.P.); (J.T.)
- Université Paul Sabatier de Toulouse, IPBS, Toulouse F-31077, France
| | - Muriel Golzio
- Centre National de la Recherche Scientifique (CNRS), Institut de Pharmacologie et de Biologie Structurale (IPBS) BP 64182, 205 route de Narbonne, Toulouse F-31077, France; E-Mails: (S.C.); (S.P.); (J.T.)
- Université Paul Sabatier de Toulouse, IPBS, Toulouse F-31077, France
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +33-561-175-811; Fax: +33-561-175-994
| |
Collapse
|
14
|
Tsuchida S, Arai Y, Kishida T, Takahashi KA, Honjo K, Terauchi R, Inoue H, Oda R, Mazda O, Kubo T. Silencing the expression of connexin 43 decreases inflammation and joint destruction in experimental arthritis. J Orthop Res 2013; 31:525-30. [PMID: 23165424 DOI: 10.1002/jor.22263] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 10/11/2012] [Indexed: 02/04/2023]
Abstract
The objective of the present study was to determine whether the expression of connexin 43 (Cx43) effected on inflammatory conditions in rat fibroblast-like synoviocytes (FLS) and on rat model of rheumatoid arthritis (RA). The expression of Cx43 in rat FLS stimulated with lipopolysaccharide (LPS) was confirmed by real-time reverse transcriptase polymerase chain reaction (RT-PCR). The effects of small-interfering RNA targeting Cx43 (siCx43) on pro-inflammatory cytokines and chemokine were assessed by real-time RT-PCR and enzyme-linked immunosorbent assay (ELISA). The therapeutic and side effects of siCx43 in a rat model of collagen-induced arthritis (CIA) were examined by in vivo electroporation method. LPS markedly enhanced Cx43 gene expression in rat FLS, with transfection of siCx43 suppressing the over-expression of pro-inflammatory cytokines and the chemokine. Treatment of CIA rats with siCx43 significantly ameliorated paw swelling, and significantly reduced histological arthritis scores and radiographic scores. In histological appearance of rat ankle joints, siCx43 treatment significantly decreased the number of tartrate-resistant acid phosphatase (TRAP)-positive (osteoclast-like) cells. These findings indicated that siCx43 had anti-inflammatory effects in rat FLS and efficiently inhibited the development of CIA. Cx43 may play an important role in the pathophysiology of RA, and may be a potential target molecule for novel RA therapies.
Collapse
Affiliation(s)
- Shinji Tsuchida
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
|
16
|
Présumey J, Salzano G, Courties G, Shires M, Ponchel F, Jorgensen C, Apparailly F, De Rosa G. PLGA microspheres encapsulating siRNA anti-TNFalpha: efficient RNAi-mediated treatment of arthritic joints. Eur J Pharm Biopharm 2012; 82:457-64. [PMID: 22922428 DOI: 10.1016/j.ejpb.2012.07.021] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 07/30/2012] [Accepted: 07/31/2012] [Indexed: 01/13/2023]
Abstract
The aim of this study was to investigate potentialities of poly(dl-lactide-co-glycolide) (PLGA) microspheres for the delivery of small interfering RNAs (siRNAs) against tumor necrosis factor α (TNF-α) to achieve prolonged and efficient inhibition of TNF-α for the treatment of rheumatoid arthritis (RA). PLGA microspheres were prepared by a modified multiple emulsion-solvent evaporation method. The formulations were characterized in terms of morphology, mean diameter and siRNAs distribution, encapsulation efficiency, and in vitro release kinetics. The efficiency of this system was then evaluated both in vitro and in vivo using the murine monocytic cell line J774 and a pre-clinical model of RA, respectively. siRNA-encapsulating PLGA microspheres were characterized by a high encapsulation efficiency and a slow and prolonged anti-TNF-α siRNAs. Our results provide evidence that, upon intra-articular administration, PLGA microspheres slowly releasing siRNAs effectively inhibited the expression of TNF-α in arthritic joints. Our system might represent an alternative strategy for the design of novel anti-rheumatic therapies based on the use of RNA interference in RA.
Collapse
Affiliation(s)
- J Présumey
- Inserm, U 844, INM, Hôpital Saint Eloi, Montpellier, France
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Komano Y, Yagi N, Onoue I, Kaneko K, Miyasaka N, Nanki T. Arthritic Joint-Targeting Small Interfering RNA-Encapsulated Liposome: Implication for Treatment Strategy for Rheumatoid Arthritis. J Pharmacol Exp Ther 2011; 340:109-13. [DOI: 10.1124/jpet.111.185884] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
18
|
Abstract
Tumor necrosis factor-α (TNF-α), a proinflammatory cytokine, plays a key role in the pathogenesis of many inflammatory diseases, including arthritis. Neutralization of this cytokine by anti-TNF-α antibodies has shown its efficacy in rheumatoid arthritis (RA) and is now widely used. Nevertheless, some patients currently treated with anti-TNF-α remain refractory or become nonresponder to these treatments. In this context, there is a need for new or complementary therapeutic strategies. In this study, we investigated in vitro and in vivo anti-inflammatory potentialities of an anti-TNF-α triplex-forming oligonucleotide (TFO), as judged from effects on two rat arthritis models. The inhibitory activity of this TFO on articular cells (synoviocytes and chondrocytes) was verified and compared to that of small interfering RNA (siRNA) in vitro. The use of the anti-TNF-α TFO as a preventive and local treatment in both acute and chronic arthritis models significantly reduced disease development. Furthermore, the TFO efficiently blocked synovitis and cartilage and bone destruction in the joints. The results presented here provide the first evidence that gene targeting by anti-TNF-α TFO modulates arthritis in vivo, thus providing proof-of-concept that it could be used as therapeutic tool for TNF-α-dependent inflammatory disorders.
Collapse
|
19
|
Mountziaris PM, Sing DC, Chew SA, Tzouanas SN, Lehman ED, Kasper FK, Mikos AG. Controlled release of anti-inflammatory siRNA from biodegradable polymeric microparticles intended for intra-articular delivery to the temporomandibular joint. Pharm Res 2010; 28:1370-84. [PMID: 21184147 DOI: 10.1007/s11095-010-0354-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Accepted: 12/14/2010] [Indexed: 01/08/2023]
Abstract
PURPOSE As the next step in the development of an intra-articular controlled release system to treat painful temporomandibular joint (TMJ) inflammation, we developed several biodegradable poly(DL-lactic-co-glycolic acid) (PLGA)-based microparticle (MP) formulations encapsulating a model anti-inflammatory small interfering RNA (siRNA) together with branched poly(ethylenimine) (PEI) as a transfecting agent. The effect of siRNA loading and N:P ratio on the release kinetics of siRNA-PEI polyplexes was determined, and the size and N:P ratio of the polyplexes released over time was characterized. METHODS Polyplex-loaded PLGA MPs were prepared using an established double emulsion technique. Increasing the pH of the release samples enabled siRNA-PEI dissociation and subsequent measurement of the release of each component over 28 days. Polyplex diameter was measured for all release samples and compared to freshly prepared siRNA-PEI under simulated physiologic conditions. RESULTS Systematic variation of siRNA loading and N:P ratio resulted in distinct siRNA and PEI release profiles. Polyplex diameter remained constant despite large variations in the relative amounts of siRNA and PEI. Excess PEI was sequestered through complexation with 500-1,000 nm diameter PLGA MP-derived particles, including small MPs and PLGA degradation products. CONCLUSIONS These PLGA MP formulations show exciting potential as the first intra-articular TMJ controlled release system.
Collapse
Affiliation(s)
- Paschalia M Mountziaris
- Department of Bioengineering, Rice University, PO Box 1892, MS 142, Houston, Texas 77251-1892, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Shi Q, Zhang XL, Dai KR, Benderdour M, Fernandes JC. siRNA therapy for cancer and non-lethal diseases such as arthritis and osteoporosis. Expert Opin Biol Ther 2010; 11:5-16. [DOI: 10.1517/14712598.2010.532483] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
21
|
Jorgensen C, Apparailly F. Prospects for gene therapy in inflammatory arthritis. Best Pract Res Clin Rheumatol 2010; 24:541-52. [DOI: 10.1016/j.berh.2010.02.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
22
|
Honjo K, Takahashi KA, Mazda O, Kishida T, Shinya M, Tokunaga D, Arai Y, Inoue A, Hiraoka N, Imanishi J, Kubo T. MDR1a/1b gene silencing enhances drug sensitivity in rat fibroblast-like synoviocytes. J Gene Med 2010; 12:219-27. [PMID: 19950109 DOI: 10.1002/jgm.1378] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Drug resistance mediated by P-glycoprotein (P-gp) is one of the major reasons for the failure of rheumatoid arthritis (RA) therapy with disease modifying anti-rheumatic drugs and glucocorticoids. In the present study, we aimed to investigate the in vitro effectiveness of small interfering RNA (siRNA) to render rat fibroblast-like synoviocytes (FLS) susceptible to drugs. We also attempted the electroporation-mediated transfer of siRNA against multidrug resistance (MDR) genes into rat knee joints. METHODS FLS were transfected with siRNAs corresponding to MDR1a and MDR1b genes. FLS were treated with dexamethasone (DEX) and lipopolysaccharide. The mRNA and protein levels of tumor necrosis factor-alpha, interleukin (IL)-6 and IL-1beta were measured. Both siRNAs were co-transduced into rat knee joints by an electroporation method and evaluated the target gene expressions in the synovium. RESULTS Each siRNA could sequence-specifically reduce the target gene expression by over 70% and effectively suppressed P-gp expression and function in the FLS. Both gene expression and protein production of the inflammatory cytokines in the cells transfected with siRNA were reduced by a greater amount compared to in control cells. The in vivo electroporation-mediated transduction of siRNA could significantly inhibit the target gene expressions. CONCLUSIONS MDR1a/1b gene silencing by siRNA could effectively inhibit P-gp in rat FLS, resulting in a significant enhancement of the anti-inflammatory effects of DEX. The in vivo siRNA transduction could successfully silence MDR gene expression in the rat synovium. These findings indicate that the siRNA targeting MDR gene could be a useful tool for treating refractory arthritis in RA.
Collapse
Affiliation(s)
- Kuniaki Honjo
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Khoury M, Courties G, Fabre S, Bouffi C, Seemayer CA, Vervoordeldonk MJ, Tak PP, Jorgensen C, Apparailly F. Adeno-associated virus type 5-mediated intraarticular administration of tumor necrosis factor small interfering RNA improves collagen-induced arthritis. ACTA ACUST UNITED AC 2010; 62:765-70. [DOI: 10.1002/art.27302] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
24
|
Zhang F, Yao Y, Hao J, Zhou R, Liu C, Gong Y, Wang DA. A dual-functioning adenoviral vector encoding both transforming growth factor-β3 and shRNA silencing type I collagen: Construction and controlled release for chondrogenesis. J Control Release 2010; 142:70-7. [DOI: 10.1016/j.jconrel.2009.09.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2009] [Revised: 09/25/2009] [Accepted: 09/29/2009] [Indexed: 11/26/2022]
|
25
|
Courties G, Presumey J, Duroux-Richard I, Jorgensen C, Apparailly F. RNA interference-based gene therapy for successful treatment of rheumatoid arthritis. Expert Opin Biol Ther 2010; 9:535-8. [PMID: 19392574 DOI: 10.1517/14712590902926089] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND RNA interference (RNAi) is a powerful endogenous process initiated by short double-stranded RNAs, which results in sequence-specific posttranscriptional gene silencing. The possibility of blocking the expression of any protein carries huge expectations for potential therapeutic applications in a wide range of diseases. For clinical development, however, the use of RNAi-based therapeutics has to overcome major obstacles, mainly targeted delivery and safety issues. OBJECTIVE/METHODS In this short review, we provide an overview of specifications for RNAi-based gene therapy in rheumatoid arthritis (RA) and discuss recent progresses in the development of efficient silencing, focusing on expression of short hairpin RNAs. RESULTS/CONCLUSIONS Combining advances in RNAi methodology with gene therapy technology opens avenues for rapid applications to RA.
Collapse
Affiliation(s)
- Gabriel Courties
- Inserm, U844, Université Montpellier 1, CHU Lapeyronie, Montpellier, France
| | | | | | | | | |
Collapse
|
26
|
Nakai N, Kishida T, Hartmann G, Katoh N, Imanishi J, Kishimoto S, Mazda O. Mitf silencing cooperates with IL-12 gene transfer to inhibit melanoma in mice. Int Immunopharmacol 2010; 10:540-5. [PMID: 20074674 DOI: 10.1016/j.intimp.2009.12.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Revised: 11/15/2009] [Accepted: 12/28/2009] [Indexed: 11/15/2022]
Abstract
Malignant melanoma is a malignant neoplasm originating from the melanocyte lineage. Microphthalmia-associated transcription factor (Mitf) is crucially involved in the melanin synthesis as well as proliferation and survival of melanocyte and melanoma. We previously showed that short interfering RNA (siRNA) that is specific for the Mitf gene (Mitf-siRNA) significantly inhibited growth of B16 melanoma after electro-transfected in vivo into preestablished tumor in mice. Here we assessed efficacy of electroporation-mediated co-transfection of Mitf-siRNA and IL-12 gene in the treatment of murine melanoma. As results, the tumor growth was more strongly inhibited by intratumor co-transfection with Mitf-siRNA and IL-12-encoding plasmid DNA than by transfection with either of the molecules alone. The co-transfection induced intratumor infiltration of CD4+ and CD8+ T cells, and hampered neoangiogenesis in the tumor. The findings suggest that the RNAi/cytokine gene combination therapy by means of electroporation may become a novel and efficacious therapeutic modality to treat neoplasms including melanoma.
Collapse
Affiliation(s)
- Noriaki Nakai
- Department of Dermatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamikyo, Kyoto 602-8566, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Nakagawa S, Arai Y, Mori H, Matsushita Y, Kubo T, Nakanishi T. Small interfering RNA targeting CD81 ameliorated arthritis in rats. Biochem Biophys Res Commun 2009; 388:467-72. [PMID: 19580788 DOI: 10.1016/j.bbrc.2009.06.150] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2009] [Accepted: 06/30/2009] [Indexed: 11/28/2022]
Abstract
CD81 belongs to a family of cell-surface protein (tetraspanin) known as one of the up-regulated elements in rheumatoid arthritis synoviocytes. In this study, the therapeutic effect of small interfering RNA targeting CD81 (siCD81) was examined by in vivo electroporation method. Treatment with siCD81 significantly ameliorated paw swelling of collagen-induced arthritic (CIA) rats. In histological examination, hypertrophy of synovium, bone erosion, and degeneration of articular cartilage were milder in rats treated with siCD81 than in the control group and the non-specific siRNA group. Expression of synoviolin, a rheumatoid regulator, was suppressed by siCD81. Thus, therapeutic intervention by targeting CD81 may be used in the treatment of rheumatoid arthritis.
Collapse
Affiliation(s)
- Shuji Nakagawa
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | | | | | | | | | | |
Collapse
|
28
|
Sakai T, Kawaguchi M, Kosuge Y. siRNA-mediated gene silencing in the salivary gland using in vivo microbubble-enhanced sonoporation. Oral Dis 2009; 15:505-11. [PMID: 19519620 DOI: 10.1111/j.1601-0825.2009.01579.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVES siRNA-induced gene silencing in the salivary gland using microbubble-enhanced sonoporation was used to develop an in vivo gene knockdown technique. METHODS siRNA targeting rat glyceraldehyde-3-phosphate dehydrogenas (GAPDH) was mixed with echo-enhanced microbubbles and reverse-injected into rat parotid glands using transdermal ultrasound. To compare direct and transdermal ultrasound efficiencies, an incision was made on the lateral neck to expose the parotid glands for direct application. The efficiency of gene suppression was determined using quantitative reverse transcription-polymerase chain reaction 24-72 h after siRNA delivery. Cytotoxicity was assessed using histological analysis. RESULTS Expression of rat GAPDH in the parotid glands was silenced 48 h after siRNA was delivered by ultrasound (frequency: 1 MHz; intensity: 2 W cm(-2); exposure time: 2 min). High-intensity ultrasound induced tissue damage and apoptotic change. Echo-enhanced microbubbles significantly improved siRNA-induced gene silencing by 10-50%. Compared with transdermal application, direct-exposure ultrasound was only slightly effective, and no significant difference in gene expression was observed. CONCLUSION The results indicate that microbubble-enhanced sonoporation can yield in vivo siRNA gene silencing in the rat parotid gland. This technique could be applied to provide gene knockdown organs for functional genomic analyses and to develop siRNA-based gene therapy.
Collapse
Affiliation(s)
- T Sakai
- Department of Pharmacology, Oral Health Science Center HRC7, Tokyo Dental College, 1-2-2 Masago, Mihama-ku, Chiba, Chiba, 261-8502, Japan.
| | | | | |
Collapse
|
29
|
Foged C, Nielsen HM, Frokjaer S. Phospholipase A2Sensitive Liposomes for Delivery of Small Interfering RNA (siRNA). J Liposome Res 2008; 17:191-6. [DOI: 10.1080/08982100701530373] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
30
|
Comparison of anti-rheumatic effects of local RNAi-based therapy in collagen induced arthritis rats using various cytokine genes as molecular targets. Mod Rheumatol 2008; 19:125-33. [PMID: 19030778 DOI: 10.1007/s10165-008-0131-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2008] [Accepted: 09/24/2008] [Indexed: 10/21/2022]
Abstract
RNA interference (RNAi) provides a powerful means of sequence-specific gene silencing. Several studies show that RNAi may provide promising strategies to treat human diseases by suppressing disease responsible genes in vivo. In locomotor diseases, the progression of collagen-induced arthritis (CIA) is suppressed by tumor necrosis factor-alpha (TNF-alpha)-specific small interfering RNA (siRNA) delivered into the joint. The aim of this study, is to compare the effects of intraarticularly administered siRNAs targeting TNF-alpha, interleukin-1beta (IL-1beta), interleukin-6 (IL-6) and receptor activator of NF-kappaB ligand (RANKL) on CIA in rats. We confirmed that the silencing effects of siRNA duplexes specific for rat TNF-alpha, IL-1beta, IL-6 and RANKL in vitro. Each siRNA was also delivered into the knee joint of CIA rats by the in vivo electroporation method 7, 10, 13 and 16 days after immunization with collagen. Local delivery of TNF-alpha or IL-1beta-specific siRNA ameliorated CIA in rats effectively at the gross morphological, radiographical and histological evaluations. Our results suggested that TNF-alpha and IL-1beta were the cytokines to be targeted in the joint for the treatment of rheumatoid arthritis. The in vivo siRNA transfection method may be useful for selection of target molecules to be silenced for treatment of joint diseases.
Collapse
|
31
|
Wang ZH, Yang ZQ, He XJ, Wang L, Li LX, Tu JB. Effects of RNAi-mediated inhibition of aggrecanase-1 and aggrecanase-2 on rat costochondral chondrocytes in vitro. Acta Pharmacol Sin 2008; 29:1215-26. [PMID: 18817627 DOI: 10.1111/j.1745-7254.2008.00856.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
AIM Failure of transplanted cartilage or allogenic chondrocytes is attributed mainly to immunological rejection and cartilage degradation. A major feature is the loss of aggrecan from the cartilage matrix, primarily due to the action of the specific proteinases aggrecanase-1 and aggrecanase-2. The aim of this in vitro study was to determine whether the specific inhibition of aggrecanase-1 and aggrecanase-2 by RNAi would mitigate aggrecan loss from cultured chondrocytes. METHODS Expression plasmid vectors of shRNA targeting aggrecanase-1 and aggrecanase-2 were constructed and transfected into cultured rattus costochondral chondrocytes. The transfected cells were induced with interleukin-1beta (IL-1beta). Gene mRNA levels were analyzed by RT-PCR. Aggrecan and collagen II content were measured by immunohistochemistry and Western blotting. RESULTS As the chondrocytes underwent dedifferentiation, aggrecanase-1 increased significantly. The specific inhibition of aggrecanase-1 and aggrecanase-2 by RNAi had no negative effect on the morphology and growth velocity of the chondrocytes. The mRNA of aggrecanase-1 and aggrecanase-2 decreased significantly. The alpha-2-macroglobulin expression level was increased by the shRNA specific for aggrecanase-1. Other genes of the chondrocytic extracellular matrix were not affected. RNAi significantly increased the aggrecan and collagen II content of chondrocytes treated with IL-1beta. CONCLUSION The results suggest that inhibition of aggrecanase-1 and aggrecanase-2 by RNAi can mitigate aggrecan degradation, without interfering with chondrocytic gene phenotype recovery. RNAi technology can be a useful tool for studying degenerative processes in cartilage.
Collapse
Affiliation(s)
- Zheng-hui Wang
- Department of Oral and Maxillofacial Plastic Surgery, Stomatological Hospital, Xi-an Jiao Tong University, Xi-an 710004, China
| | | | | | | | | | | |
Collapse
|
32
|
Mountziaris PM, Kramer PR, Mikos AG. Emerging intra-articular drug delivery systems for the temporomandibular joint. Methods 2008; 47:134-40. [PMID: 18835358 DOI: 10.1016/j.ymeth.2008.09.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2008] [Revised: 08/31/2008] [Accepted: 09/05/2008] [Indexed: 12/12/2022] Open
Abstract
Temporomandibular joint (TMJ) disorders are a heterogeneous group of diseases that cause progressive joint degeneration leading to chronic pain and reduced quality of life. Both effective pain reduction and restoration of TMJ function remain unmet challenges. Intra-articular injections of corticosteroids and hyaluronic acid are currently used to treat chronic pain, but these methods require multiple injections that increase the risk of iatrogenic joint damage and other complications. The small and emerging field of TMJ tissue engineering aims to reduce pain and disability through novel strategies that induce joint tissue regeneration. Development of methods for sustained, intra-articular release of growth factors and other pro-regenerative signals will be critical for the success of TMJ tissue engineering strategies. This review discusses methods of intra-articular drug delivery to the TMJ, as well as emerging injectable controlled release systems with potential to improve TMJ drug delivery, to encourage further research in the development of sustained release systems for both long-term pain management and to enhance tissue engineering strategies for TMJ regeneration.
Collapse
Affiliation(s)
- Paschalia M Mountziaris
- Department of Bioengineering, Rice University, P.O. Box 1892, MS 142, Houston, TX 77251-1892, USA
| | | | | |
Collapse
|
33
|
Khoury M, Escriou V, Courties G, Galy A, Yao R, Largeau C, Scherman D, Jorgensen C, Apparailly F. Efficient suppression of murine arthritis by combined anticytokine small interfering RNA lipoplexes. ACTA ACUST UNITED AC 2008; 58:2356-67. [DOI: 10.1002/art.23660] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
34
|
Abstract
The successful use of small interfering RNAs (siRNAs) for therapeutic purposes requires safe and efficient delivery to specific cells and tissues. In this study, we demonstrate cell type-specific delivery of anti-human immunodeficiency virus (anti-HIV) siRNAs through fusion to an anti-gp120 aptamer. The envelope glycoprotein is expressed on the surface of HIV-1-infected cells, allowing binding and internalization of the aptamer-siRNA chimeric molecules. We demonstrate that the anti-gp120 aptamer-siRNA chimera is specifically taken up by cells expressing HIV-1 gp120, and that the appended siRNA is processed by Dicer; this releases an anti-tat/rev siRNA which, in turn, inhibits HIV replication. We show for the first time a dual functioning aptamer-siRNA chimera in which both the aptamer and the siRNA portions have potent anti-HIV activities. We also show that gp120 expressed on the surface of HIV-infected cells can be used for aptamer-mediated delivery of anti-HIV siRNAs.
Collapse
|
35
|
Buyens K, Lucas B, Raemdonck K, Braeckmans K, Vercammen J, Hendrix J, Engelborghs Y, De Smedt SC, Sanders NN. A fast and sensitive method for measuring the integrity of siRNA-carrier complexes in full human serum. J Control Release 2007; 126:67-76. [PMID: 18068258 DOI: 10.1016/j.jconrel.2007.10.024] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Revised: 10/25/2007] [Accepted: 10/29/2007] [Indexed: 12/18/2022]
Abstract
SiRNA based therapeutics are currently under investigation for treatment of cancer and viral infections. Upon intravenous administration, the nanoscopic delivery systems which carry the siRNA need to be stable in serum, an aspect which is often overlooked in numerous publications on siRNA delivery systems. Techniques currently available for studying the dissociation of siRNA-liposome complexes are time consuming and incompatible with full serum. We therefore developed a fluorescence fluctuation spectroscopy (FFS) based method which allows to monitor the integrity of siRNA-carrier complexes. The method can very rapidly provide quantitative information on the complex integrity in biological media, like full human serum, and at very low siRNA concentrations (approximately 20 nM siRNA). Information on the integrity of intravenously injected siRNA nanoparticles in serum is crucial. Consequently, the FFS method reported in this work may find broad applicability in the field of siRNA-carrier design.
Collapse
Affiliation(s)
- Kevin Buyens
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Saito M, Mazda O, Takahashi KA, Arai Y, Kishida T, Shin-Ya M, Inoue A, Tonomura H, Sakao K, Morihara T, Imanishi J, Kawata M, Kubo T. Sonoporation mediated transduction of pDNA/siRNA into joint synovium in vivo. J Orthop Res 2007; 25:1308-16. [PMID: 17549706 DOI: 10.1002/jor.20392] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The purpose of this study was to investigate the usefulness of sonoporation method on in vivo transduction of plasmid DNA (pDNA) and small interfering RNA (siRNA) into joint tissue. pGEG.GL3 plasmid was mixed with microbubble and injected into knee joints of rats. Ultrasound sonication was performed percutaneously. Three days after injection, GL3 expression of synovial tissue was determined by luciferase assay and RT-PCR. siRNA specific for GL3 (siGL3) or nonspecific siRNA were mixed with pGEG.GL3 plasmid and transduced by sonoporation. siRNA specific for EGFP (siEGFP) was transduced into the knee joints of EGFP transgenic rats, and gene silencing effects for endogenous gene were examined. To determine the localization of transduced siRNA, fluorescently labeled siRNA was transduced into joints. The expression of GL3 in the synovium was significantly enhanced by sonoporation. The gene expression was only seen in the synovium of the knee joint. The expression of GL3 was remarkably suppressed by co-transduction of siGL3, but not suppressed by nonspecific siRNA. siEGFP transduced by sonoporation attenuated green fluorescence on the surface layer of synovium of EGFP transgenic rats. The fluorescently labeled siRNA was seen in the synovium around the patella, femur, and tibia. Sonoporation is examined as a recent, novel, gene transduction method, and the advantage of this technique is minimal invasiveness. In this study, we showed that pDNA/siRNA can be transduced specifically into the joint synovium using sonoporation. The present method may be useful in nucleic acid therapy for joint disorders.
Collapse
Affiliation(s)
- Masazumi Saito
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Gould DJ, Chernajovsky Y. Novel delivery methods to achieve immunomodulation. Curr Opin Pharmacol 2007; 7:445-50. [PMID: 17611159 PMCID: PMC2679984 DOI: 10.1016/j.coph.2007.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2007] [Revised: 05/22/2007] [Accepted: 05/22/2007] [Indexed: 01/13/2023]
Abstract
Immunomodulation in infectious diseases, cancer, cardiovascular disease and autoimmunity can now be targeted by sophisticated protein design, altering cellular responses by increasing therapeutic cell numbers ex vivo and then reimplanting, or altering cell function by gene transfer of cells ex vivo. In the last year, vaccination has been applied to modulate responses to autoantigens, allergens, viral or cancer antigens. The application of these technologies has entered the clinical arena and is having a positive impact on the treatment and prevention of human diseases.
Collapse
|
38
|
Lee UJ, Choung SR, Prakash KVB, Lee EJ, Lee MY, Kim YJ, Han CW, Choi YC. Dual knockdown of p65 and p50 subunits of NF-kappaB by siRNA inhibits the induction of inflammatory cytokines and significantly enhance apoptosis in human primary synoviocytes treated with tumor necrosis factor-alpha. Mol Biol Rep 2007; 35:291-8. [PMID: 17530443 DOI: 10.1007/s11033-007-9084-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2007] [Accepted: 04/09/2007] [Indexed: 01/04/2023]
Abstract
In order to develop an anti-NF-kappaB siRNA as a novel class of anti-inflammatory drug, we have isolated a highly efficient siRNA targeting the p65 (RelA) subunit of NF-kappaB, hereafter named REL1096. To determine whether down-regulation of p65 by REL1096 can block the induction of inflammatory cytokines after treatment with tumor necrosis factor-alpha (TNF-alpha), human primary fibroblast-like synoviocytes were isolated from patients with rheumatoid arthritis. When treated with REL1096, the TNF-mediated induction of downstream target genes such as inflammatory cytokines, chemokines, and anti-apoptosis genes was drastically inhibited. To enhance the inhibitory effect of REL1096, cells were treated with siRNA targeting the p50 subunit of NF-kappaB together with REL1096. In addition to effective downregulation of inflammatory cytokines, knockdown of both p65 and p50 resulted in much more extensive apoptosis when compared to cells treated with either REL1096 or p50-siRNA alone. Thus, our results provide evidence for the potential use of siRNA targeting NF-kappaB as an effective means to treat rheumatoid arthritis. In addition to effective amelioration of synovial inflammation by downregulation of inflammatory cytokines, increased apoptosis by dual knockdown of p65 and p50 may prove advantageous in preventing invasiveness and destructiveness of hyperplastic synoviocytes.
Collapse
Affiliation(s)
- Ui Jin Lee
- Daejeon St. Mary's Hospital, College of Medicine, Catholic University of Korea, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Kim WJ, Chang CW, Lee M, Kim SW. Efficient siRNA delivery using water soluble lipopolymer for anti-angiogenic gene therapy. J Control Release 2007; 118:357-63. [PMID: 17313987 DOI: 10.1016/j.jconrel.2006.12.026] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2006] [Revised: 12/20/2006] [Accepted: 12/28/2006] [Indexed: 12/27/2022]
Abstract
We examined the potential application of a non-viral gene carrier, water soluble lipopolymer (WSLP) for delivering siRNA targeting vascular endothelial growth factor (VEGF) in vitro and in vivo. WSLP was complexed with siRNA designed to inhibit human VEGF expression or scrambled siRNA as a control. WSLP readily formed nano-sized complexes ( approximately 100 nm) with siRNA and protected siRNAs from enzymatic degradation in serum conditioned media. WSLP/siRNA complexes were transfected in PC-3 cells derived from human prostate adenocarcinomas and, then the siRNA delivery efficiency of the complexes was evaluated by VEGF production inhibition assay. VEGF production was efficiently inhibited by the WSLP/siRNA complexes, while complexes of WSLP with scrambled siRNA did not show this inhibitory effect. WSLP/siRNA complexes reduced the VEGF production by 40% when compared to unmodified branched polyethylenimine (bPEI, MW=1800). Moreover, WSLP/siRNA complexes reduced tumor volume by 55% at 21 days, and by 65% at 28 days when compared to controls. These results indicate that WSLP has potential as a siRNA delivering agent and can be applied for anti-angiogenic tumor therapy.
Collapse
Affiliation(s)
- Won Jong Kim
- Center for Controlled Chemical Delivery, Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112-5820, USA
| | | | | | | |
Collapse
|
40
|
Song RH, Tortorella MD, Malfait AM, Alston JT, Yang Z, Arner EC, Griggs DW. Aggrecan degradation in human articular cartilage explants is mediated by both ADAMTS-4 and ADAMTS-5. ACTA ACUST UNITED AC 2007; 56:575-85. [PMID: 17265492 DOI: 10.1002/art.22334] [Citation(s) in RCA: 301] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Recent published studies have shown that cartilage from ADAMTS-5-knockout mice, but not ADAMTS-4- or ADAMTS-1-knockout mice, is significantly protected from degradation. The present study was undertaken to evaluate the respective roles of these enzymes in human cartilage breakdown, using a small interfering RNA (siRNA) approach to assess the effects of inhibition of each enzyme in normal and osteoarthritic (OA) explants. METHODS The activities of siRNA specifically targeting ADAMTS-1, -4, and -5 were assessed by transfection into primary human chondrocytes and cultured human cartilage explants. At 24 hours, a cytokine stimulus was applied to normal, but not OA, samples to initiate a catabolic response. At designated times, total RNA was isolated and gene expression was measured by quantitative real-time reverse transcription-polymerase chain reaction. Aggrecan release and aggrecanase-generated neoepitope formation were determined by dye binding analysis and Western blotting, respectively. RESULTS Human chondrocytes and explants were efficiently transfected with siRNA that specifically decreased the expression of each targeted gene. Suppression of ADAMTS-4 and ADAMTS-5, individually or in combination, attenuated the degradation of aggrecan in cytokine-stimulated normal cartilage. A reduction in aggrecan degradation was also observed following siRNA-mediated knockdown of either gene in unstimulated OA cartilage. In contrast, knockdown of ADAMTS-1 failed to inhibit aggrecan loss. CONCLUSION Despite the apparent dominant role of ADAMTS-5 in genetically modified mice, our data suggest that both ADAMTS-4 and ADAMTS-5 contribute to the structural damage that characterizes human OA.
Collapse
Affiliation(s)
- Ruo-Hua Song
- Pfizer Global Research and Development, St Louis, MO 63017, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Golzio M, Mazzolini L, Ledoux A, Paganin A, Izard M, Hellaudais L, Bieth A, Pillaire MJ, Cazaux C, Hoffmann JS, Couderc B, Teissié J. In vivo gene silencing in solid tumors by targeted electrically mediated siRNA delivery. Gene Ther 2007; 14:752-9. [PMID: 17344906 DOI: 10.1038/sj.gt.3302920] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
RNA interference (RNAi)-mediated gene silencing approaches appear very promising for therapies based on the targeted inhibition of disease-relevant genes. The major hurdle to the therapeutic development of RNAi strategies remains, however, the efficient delivery of the RNAi-inducing molecules, the short interfering RNAs (siRNAs) and short hairpin RNAs (shRNAs), to the target tissue. With respect to cancer treatment the development of efficient delivery methods into solid tumors appears as a critical issue. However, very few studies have addressed this problem. In this study we have investigated the contribution of electrically mediated delivery of siRNA into murine tumors stably expressing an enhanced green fluorescent protein (EGFP) target reporter gene. The silencing of EGFP gene expression was quantified over time by fluorescence imaging in the living animal. Our study indicates that electric field can be used as an efficient method for siRNA delivery and associated gene silencing into cells of solid tumors in vivo.
Collapse
|
42
|
Affiliation(s)
- Loree C Heller
- Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | | |
Collapse
|
43
|
Nakai N, Kishida T, Shin-Ya M, Imanishi J, Ueda Y, Kishimoto S, Mazda O. Therapeutic RNA interference of malignant melanoma by electrotransfer of small interfering RNA targeting Mitf. Gene Ther 2006; 14:357-65. [PMID: 17024102 DOI: 10.1038/sj.gt.3302868] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Microphthalmia-associated transcription factor (Mitf) is critically involved in melanin synthesis as well as differentiation of cells of the melanocytic lineage. Some earlier studies suggested that Mitf is also essential in the survival of melanoma cells, but this notion remains controversial. We synthesized short interfering RNA (siRNA) duplexes corresponding to the mitf sequence and transfected them into B16 melanoma. Lipid-mediated transfection in vitro of Mitf-specific siRNA resulted in specific downregulation of Mitf and of the tyrosinase that is a transcriptional target of Mitf. This treatment also remarkably reduced the viability of melanoma cells by inducing apoptosis. To examine the potential feasibility of RNAi therapy against melanoma, B16 cells were subcutaneously injected into syngenic mice and siRNA was transfected into the pre-established tumor by means of electroporation. The Mitf-specific siRNA drastically reduced outgrowth of subcutaneous melanoma, while nonspecific siRNA failed to affect tumor progression. Terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling-based analysis of tumor specimens demonstrated that the tumor cells transfected with Mitf-siRNA effectively underwent apoptosis in vivo. The present results indicate that Mitf plays important roles in melanoma survival. Intratumor electrotransfer of Mitf-specific siRNA may provide a powerful strategy for therapeutic intervention of malignant melanoma.
Collapse
Affiliation(s)
- N Nakai
- Department of Dermatology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | | | | | | | | | | |
Collapse
|
44
|
|
45
|
Behlke MA. Progress towards in vivo use of siRNAs. Mol Ther 2006; 13:644-70. [PMID: 16481219 PMCID: PMC7106286 DOI: 10.1016/j.ymthe.2006.01.001] [Citation(s) in RCA: 325] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2005] [Revised: 01/11/2006] [Accepted: 01/11/2006] [Indexed: 01/28/2023] Open
Abstract
RNA interference (RNAi) has become the method of choice to suppress gene expression in vitro. It is also emerging as a powerful tool for in vivo research with over 90 studies published using synthetic small interfering RNAs in mammals. These reports demonstrate the potential for use of synthetic small interfering RNAs (siRNAs) as therapeutic agents, especially in the areas of cancer and viral infection. The number of reports using siRNAs for functional genomics applications, for validation of targets for small-molecule drug development programs, and to address questions of basic biology will rapidly grow as methods and protocols for use in animals become more established. This review will first discuss aspects of RNAi biochemistry and biology that impact in vivo use, especially as relates to experimental design, and will then provide an overview of published work with a focus on methodology.
Collapse
Affiliation(s)
- Mark A Behlke
- Integrated DNA Technologies, Inc., Coralville, IA 52241, USA.
| |
Collapse
|
46
|
Khoury M, Louis-Plence P, Escriou V, Noel D, Largeau C, Cantos C, Scherman D, Jorgensen C, Apparailly F. Efficient new cationic liposome formulation for systemic delivery of small interfering RNA silencing tumor necrosis factor α in experimental arthritis. ACTA ACUST UNITED AC 2006; 54:1867-77. [PMID: 16729293 DOI: 10.1002/art.21876] [Citation(s) in RCA: 149] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Tumor necrosis factor alpha (TNFalpha) is among the most prominent cytokines in rheumatoid arthritis (RA) and is secreted mainly by macrophages. A direct method for restoring the immunologic balance in RA is use of small interfering RNA (siRNA) for silencing the TNFalpha transcript. The aim of this study was to determine the therapeutic effect of systemic administration of TNFalpha siRNA in an experimental model of RA, optimizing its delivery using new liposome formulations. METHODS Murine macrophages were transfected with siRNA targeting TNFalpha, and expression was measured. The therapeutic effect in collagen-induced arthritis (CIA) was assessed after intravenous delivery of TNFalpha siRNA. Delivery was optimized using a carrier DNA for complexation with the cationic liposome RPR209120/DOPE. Levels of TNFalpha and other cytokines were measured in sera and joint tissue-conditioned media. Biodistribution was determined using a fluorescent siRNA. RESULTS In vitro, TNFalpha siRNA efficiently and specifically modulated the expression of TNFalpha at both the messenger RNA and protein levels. In vivo, complete cure of CIA was observed when TNFalpha siRNA was administered weekly, complexed with the liposome and combined with carrier DNA. Inhibition (50-70%) of articular and systemic TNFalpha secretion was detected in the siRNA-injected groups, which correlated with a decrease in the levels of interleukin-6 and monocyte chemotactic protein 1. The main organs targeted by siRNA were the liver and spleen; the addition of liposome RPR209120 and carrier DNA significantly increased organ uptake. CONCLUSION We demonstrated the efficiency of systemic delivery of siRNA designed to silence TNFalpha in CIA, using a liposome carrier system as a way to address the methodologic limitations in vivo.
Collapse
Affiliation(s)
- Maroun Khoury
- INSERM U475 and University of Montpellier I, Montpellier, France.
| | | | | | | | | | | | | | | | | |
Collapse
|