1
|
Moon TK, Kang IK, Lee KJ, Kim JH, Kim HJ, Han AR, Woo HN, Lee JY, Choi JS, Park K, Lee H. mTOR downregulation promotes anti-inflammatory responses via the CCL3-CCR5 axis in hypoxic retinopathy. Mol Ther Methods Clin Dev 2025; 33:101404. [PMID: 39897639 PMCID: PMC11787640 DOI: 10.1016/j.omtm.2024.101404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 12/28/2024] [Indexed: 02/04/2025]
Abstract
Hypoxic retinopathies, including diabetic retinopathy, are major contributors to vision impairment, mainly due to accelerated angiogenesis and inflammation. Previously, we demonstrated that AAV2-shmTOR, effective across distinct species, holds therapeutic promise by modulating the activated mTOR pathway, yet its mechanisms for reducing inflammation remain largely unexplored. To investigate AAV2-shmTOR's impact on atypical inflammation in these conditions, we employed an in vivo model of oxygen-induced retinopathy and an in vitro model using rMC1 Müller cells. AAV2-shmTOR notably decreased mTOR expression in rMC1 cells under hypoxic conditions, as verified by co-staining for mTOR and glial fibrillary acidic protein (GFAP). It effectively interrupted the activation of mTOR signaling triggered by hypoxia. It diminished the secretion of CCL3 from rMC1 cells, consequently reducing microglial migration in response to conditioned media from AAV2-shmTOR-treated rMC1 cells. Notably, the virus lowered CCL3 expression in Müller cells and reduced the presence of CCR5-positive microglia in vivo, indicating its effectiveness in targeted inflammation management via the CCL3-CCR5 pathway. These findings thus highlight the potential of AAV2-shmTOR to exert anti-inflammatory effects by influencing the mTOR and subsequent CCL3-CCR5 pathways in hypoxic retinopathies, presenting a novel therapeutic approach for retinal diseases marked by hypoxia-driven inflammation.
Collapse
Affiliation(s)
- Tae Kwon Moon
- Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Microbiology, University of Ulsan, College of Medicine, Seoul, Republic of Korea
| | - Im Kyeung Kang
- Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Microbiology, University of Ulsan, College of Medicine, Seoul, Republic of Korea
| | - Kyoung Jin Lee
- Department of Microbiology, University of Ulsan, College of Medicine, Seoul, Republic of Korea
- Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ji Hyun Kim
- Department of Microbiology, University of Ulsan, College of Medicine, Seoul, Republic of Korea
- Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hee Jong Kim
- Institute of New Drug Development Research, CdmoGen Co., Ltd., Seoul 05854, Republic of Korea
| | - A. Reum Han
- Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Biochemistry & Molecular Biology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ha-Na Woo
- Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Biochemistry & Molecular Biology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Joo Yong Lee
- Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Ophthalmology, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, Republic of Korea
| | - Jun-Sub Choi
- Institute of New Drug Development Research, CdmoGen Co., Ltd., Seoul 05854, Republic of Korea
| | - Keerang Park
- Institute of New Drug Development Research, CdmoGen Co., Ltd., Seoul 05854, Republic of Korea
| | - Heuiran Lee
- Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Microbiology, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, Republic of Korea
| |
Collapse
|
2
|
Chang H, Lee KJ, Park M, Woo HN, Kim JH, Kang IK, Park H, Chon CH, Lee H, Jung HH. Cross-species RNAi therapy via AAV delivery alleviates neuropathic pain by targeting GCH1. Neurotherapeutics 2025; 22:e00511. [PMID: 39674763 PMCID: PMC12014335 DOI: 10.1016/j.neurot.2024.e00511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/27/2024] [Accepted: 12/06/2024] [Indexed: 12/16/2024] Open
Abstract
Tetrahydrobiopterin (BH4) expression is normally strictly controlled; however, its intracellular levels increase considerably following nerve damage. GTP cyclohydrolase I (GCH1) plays a crucial role in regulating BH4 concentration, with an upregulation observed in the dorsal root ganglion in cases of neuropathic pain. In this study, we aimed to develop and evaluate the clinical potential of an RNA interference-based adeno-associated virus (AAV) targeting GCH1 across various species to decrease BH4 levels and, consequently, alleviate neuropathic pain symptoms. We identified universal small-interfering RNA sequences effective across species and developed an AAV-u-shRNA that successfully suppressed GCH1 expression with minimal off-target effects. Male Sprague Dawley rats were divided into four groups: normal, spared nerve injury, AAV-shCON, and AAV-u-shGCH1. The rats were sacrificed on post-injection day 28 to collect blood for BH4 level assessment. The AAV-u-shGCH1 group demonstrated remarkable improvement in the mechanical withdrawal threshold by PID 28, significantly outperforming the normal, spared nerve injury, and AAV-shCON groups. Plasma BH4 levels confirmed that AAV-u-shGCH1 effectively reduced neuropathic pain by inhibiting BH4 synthesis in vivo, introducing a novel, multispecies-compatible therapeutic strategy. Our results suggest that a single application of AAV-u-shGCH1 could offer a viable solution for neuropathic pain relief.
Collapse
Affiliation(s)
- Heesue Chang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyoung Jin Lee
- Department of Microbiology, University of Ulsan College of Medicine, Seoul, Republic of Korea; Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Minkyung Park
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), and Research Foundation for Mental Hygiene, Inc. (RFMH), New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA
| | - Ha-Na Woo
- Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea; Department of Biochemistry & Molecular Biology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ji Hyun Kim
- Department of Microbiology, University of Ulsan College of Medicine, Seoul, Republic of Korea; Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Im Kyeung Kang
- Department of Microbiology, University of Ulsan College of Medicine, Seoul, Republic of Korea; Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | | | | - Heuiran Lee
- Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea; Department of Microbiology, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, Republic of Korea.
| | - Hyun Ho Jung
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Neurosurgery, Brain Research institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Park M, Woo HN, Koh CS, Chang H, Kim JH, Park K, Chang JW, Lee H, Jung HH. A Single Injection of rAAV-shmTOR in Peripheral Nerve Persistently Attenuates Nerve Injury-Induced Mechanical Allodynia. Int J Mol Sci 2023; 24:15918. [PMID: 37958901 PMCID: PMC10649356 DOI: 10.3390/ijms242115918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Activation of mammalian target of rapamycin (mTOR) has been known as one of the contributing factors in nociceptive sensitization after peripheral injury. Its activation followed by the phosphorylation of downstream effectors causes hyperexcitability of primary sensory neurons in the dorsal root ganglion. We investigated whether a single injection of rAAV-shmTOR would effectively downregulate both complexes of mTOR in the long-term and glial activation as well. Male SD rats were categorized into shmTOR (n = 29), shCON (n = 23), SNI (n = 13), and Normal (n = 8) groups. Treatment groups were injected with rAAV-shmTOR or rAAV-shCON, respectively. DRG tissues and sciatic nerve were harvested for Western blot and immunohistochemical analyses. Peripheral sensitization was gradually attenuated in the shmTOR group, and it reached a peak on PID 21. Western blot analysis showed that both p-mTORC1 and p-mTORC2 were downregulated in the DRG compared to shCON and SNI groups. We also found decreased expression of phosphorylated p38 and microglial activation in the DRG. We first attempted a therapeutic strategy for neuropathic pain with a low dose of AAV injection by interfering with the mTOR signaling pathway, suggesting its potential application in pain treatment.
Collapse
Affiliation(s)
- Minkyung Park
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.P.); (C.S.K.); (H.C.); (J.W.C.)
- Brain Korea 21 PLUS Project for Medical Science and Brain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Ha-Na Woo
- Department of Biochemistry & Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea;
- Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea;
| | - Chin Su Koh
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.P.); (C.S.K.); (H.C.); (J.W.C.)
| | - Heesue Chang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.P.); (C.S.K.); (H.C.); (J.W.C.)
| | - Ji Hyun Kim
- Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea;
- Department of Microbiology, Asan Medical Center, College of Medicine, University of Ulsan, Seoul 05505, Republic of Korea
| | - Keerang Park
- Cedmogen Co., Ltd., Cheongju 28644, Republic of Korea;
| | - Jin Woo Chang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.P.); (C.S.K.); (H.C.); (J.W.C.)
- Brain Korea 21 PLUS Project for Medical Science and Brain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Heuiran Lee
- Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea;
- Department of Microbiology, Asan Medical Center, College of Medicine, University of Ulsan, Seoul 05505, Republic of Korea
| | - Hyun Ho Jung
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.P.); (C.S.K.); (H.C.); (J.W.C.)
| |
Collapse
|
4
|
mTOR inhibition as a novel gene therapeutic strategy for diabetic retinopathy. PLoS One 2022; 17:e0269951. [PMID: 35709240 PMCID: PMC9202865 DOI: 10.1371/journal.pone.0269951] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 05/31/2022] [Indexed: 11/22/2022] Open
Abstract
In addition to laser photocoagulation, therapeutic interventions for diabetic retinopathy (DR) have heretofore consisted of anti-VEGF drugs, which, besides drawbacks inherent to the treatments themselves, are limited in scope and may not fully address the condition’s complex pathophysiology. This is because DR is a multifactorial condition, meaning a gene therapy focused on a target with broader effects, such as the mechanistic target of rapamycin (mTOR), may prove to be the solution in overcoming these concerns. Having previously demonstrated the potential of a mTOR-inhibiting shRNA packaged in a recombinant adeno-associated virus to address a variety of angiogenic retinal diseases, here we explore the effects of rAAV2-shmTOR-SD in a streptozotocin-induced diabetic mouse model. Delivered via intravitreal injection, the therapeutic efficacy of the virus vector upon early DR processes was examined. rAAV2-shmTOR-SD effectively transduced mouse retinas and therein downregulated mTOR expression, which was elevated in sham-treated and control shRNA-injected (rAAV2-shCon-SD) control groups. mTOR inhibition additionally led to marked reductions in pericyte loss, acellular capillary formation, vascular permeability, and retinal cell layer thinning, processes that contribute to DR progression. Immunohistochemistry showed that rAAV2-shmTOR-SD decreased ganglion cell loss and pathogenic Müller cell activation and proliferation, while also having anti-apoptotic activity, with these effects suggesting the therapeutic virus vector may be neuroprotective. Taken together, these results build upon our previous work to demonstrate the broad ability of rAAV2-shmTOR-SD to address aspects of DR pathophysiology further evidencing its potential as a human gene therapeutic strategy for DR.
Collapse
|
5
|
Lee SHS, Chang H, Kim JH, Kim HJ, Choi JS, Chung S, Woo HN, Lee KJ, Park K, Lee JY, Lee H. Inhibition of mTOR via an AAV-Delivered shRNA Tested in a Rat OIR Model as a Potential Antiangiogenic Gene Therapy. Invest Ophthalmol Vis Sci 2020; 61:45. [PMID: 32106292 PMCID: PMC7329967 DOI: 10.1167/iovs.61.2.45] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Purpose Recent studies have shown that inhibitors of the mechanistic target of rapamycin (mTOR) play important roles in proliferating endothelial cells within the retinal vasculature. Here we explore the effects of inhibiting mTOR as a potential gene therapeutic against pathological retinal angiogenesis in a rat model of oxygen-induced retinopathy (OIR). Methods Sprague-Dawley pups were used to generate the OIR model, with a recombinant adeno-associated virus expressing an shRNA (rAAV2-shmTOR-GFP) being administered via intravitreal injection on returning the rats to normoxia, with appropriate controls. Immunohistochemistry and TUNEL assays, as well as fluorescein angiography, were performed on transverse retinal sections and flat mounts, respectively, to determine the in vivo effects of mTOR inhibition. Results Compared with normal control rats, as well as OIR model animals that were either untreated (20.95 ± 6.85), mock-treated (14.50 ± 2.47), or injected with a control short hairpin RNA (shRNA)-containing virus vector (16.64 ± 4.92), rAAV2-shmTOR-GFP (4.28 ± 2.86, P = 0.00103) treatment resulted in dramatically reduced neovascularization as a percentage of total retinal area. These results mirrored quantifications of retinal avascular area and vessel tortuosity, with rAAV2-shmTOR-GFP exhibiting significantly greater therapeutic efficacy than the other treatments. The virus vector was additionally shown to reduce inflammatory cell infiltration into retinal tissue and possess antiapoptotic properties, both these processes having been implicated in the pathophysiology of angiogenic retinal disorders. Conclusions Taken together, these results demonstrate the strong promise of rAAV2-shmTOR-GFP as an effective and convenient gene therapy for the treatment of neovascular retinal diseases.
Collapse
|
6
|
Lee SHS, Chang H, Kim HJ, Choi JS, Kim J, Kim JH, Woo HN, Nah SK, Jung SJ, Lee JY, Park K, Park TK, Lee H. Effects of Stuffer DNA on the Suppression of Choroidal Neovascularization by a rAAV Expressing a mTOR-Inhibiting shRNA. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 14:171-179. [PMID: 31380463 PMCID: PMC6661460 DOI: 10.1016/j.omtm.2019.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/21/2019] [Indexed: 11/19/2022]
Abstract
Choroidal neovascularization (CNV) is the defining characteristic of the wet subtype of age-related macular degeneration (AMD), which is a rapidly growing global health problem. Previously, we had demonstrated the therapeutic potential of gene therapy against CNV using short hairpin RNA (shRNA) delivered via recombinant adeno-associated virus (rAAV), which abrogates mammalian-to-mechanistic (mTOR) activity in a novel manner by simultaneously inhibiting both mTOR complexes. Both the target and use of gene therapy represent a novel treatment modality against AMD. Here, the xenogeneic GFP gene used as a reporter in previous studies was removed from the virus vector to further develop the therapeutic for clinical trials. Instead, a stuffer DNA derived from the 3′ UTR of the human UBE3A gene was used to ensure optimal viral genome size for efficient rAAV assembly. The virus vector containing the stuffer DNA, rAAV2-shmTOR-SD, positively compares to one encoding the shRNA and a GFP expression cassette in terms of reducing CNV in a laser-induced mouse model, as determined by fundus fluorescein angiography. These results were confirmed via immunohistochemistry using anti-CD31, while a TUNEL assay showed that rAAV2-shmTOR-SD possesses anti-apoptotic properties as well. The qualities exhibited by rAAV2-shmTOR-SD demonstrate its potential as a human gene therapeutic for the treatment of wet AMD.
Collapse
Affiliation(s)
- Steven Hyun Seung Lee
- Department of Microbiology, College of Medicine, University of Ulsan, Seoul 05505, Korea
- Bio-Medical Institute of Technology, College of Medicine, University of Ulsan, Seoul 05505, Korea
| | - HeeSoon Chang
- CuroGene Life Sciences Co., Ltd., Cheongju 28578, Korea
| | - Hee Jong Kim
- CuroGene Life Sciences Co., Ltd., Cheongju 28578, Korea
| | - Jun-Sub Choi
- CuroGene Life Sciences Co., Ltd., Cheongju 28578, Korea
| | - Jin Kim
- CuroGene Life Sciences Co., Ltd., Cheongju 28578, Korea
| | - Ji Hyun Kim
- Department of Microbiology, College of Medicine, University of Ulsan, Seoul 05505, Korea
- Bio-Medical Institute of Technology, College of Medicine, University of Ulsan, Seoul 05505, Korea
| | - Ha-Na Woo
- Department of Microbiology, College of Medicine, University of Ulsan, Seoul 05505, Korea
- Bio-Medical Institute of Technology, College of Medicine, University of Ulsan, Seoul 05505, Korea
| | - Seung Kwan Nah
- Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, Bucheon 14584, Korea
| | - Sang Joon Jung
- Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, Bucheon 14584, Korea
| | - Joo Yong Lee
- Bio-Medical Institute of Technology, College of Medicine, University of Ulsan, Seoul 05505, Korea
- Department of Ophthalmology, University of Ulsan, College of Medicine, Seoul 05505, Korea
- Asan Medical Center, University of Ulsan, College of Medicine, Seoul 05505, Korea
| | - Keerang Park
- Department of Biopharmacy, Chungbuk Health & Science University, Cheongju 28150, Korea
| | - Tae Kwann Park
- Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, Bucheon 14584, Korea
- Department of Ophthalmology, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea
- Corresponding author: Tae Kwann Park, MD, PhD, Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, 170, Jomaru-ro, Wonmi-gu, Bucheon 14584, Korea.
| | - Heuiran Lee
- Department of Microbiology, College of Medicine, University of Ulsan, Seoul 05505, Korea
- Bio-Medical Institute of Technology, College of Medicine, University of Ulsan, Seoul 05505, Korea
- Corresponding author: Heuiran Lee, PhD, Department of Microbiology and Bio-Medical Institute of Technology, College of Medicine, University of Ulsan, 88 Olympic-ro 43-gil Songpa-gu, Seoul 05505, Korea.
| |
Collapse
|
7
|
Lee S, Kang IK, Kim JH, Jung BK, Park K, Chang H, Lee JY, Lee H. Relationship Between Neutralizing Antibodies Against Adeno-Associated Virus in the Vitreous and Serum: Effects on Retinal Gene Therapy. Transl Vis Sci Technol 2019; 8:14. [PMID: 31016068 PMCID: PMC6467092 DOI: 10.1167/tvst.8.2.14] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/26/2018] [Indexed: 11/24/2022] Open
Abstract
Purpose We determine the prevalence of neutralizing antibodies (NAbs) to adeno-associated virus (AAV) in the vitreous humor and serum of patients with vitreoretinal diseases and investigate the relationship between NAb titers in the vitreous humor and serum. Methods We analyzed NAbs to AAV serotypes 2, 5, 8, and 9 via in vitro neutralization in the vitreous humor and serum from 32 patients requiring vitrectomy for vitreoretinal diseases. The blood-retinal barrier (BRB) was evaluated for integrity based on preoperative examinations, with vitreous hemorrhage (VH) on funduscopy or dye leakage on fluorescein angiography observed indicating disruption. Results NAb levels were much lower in the vitreous humor than in the serum regardless of serotype. Patients with VH had higher levels of NAbs in the vitreous humor than those without VH. The NAb ratio (ratio between NAb titers in the serum and vitreous humor) was much lower in patients with epiretinal membrane with than in those without leakage. A significantly lower NAb ratio was noticed in patients with than in those without BRB disruptions. Conclusions The NAb ratio between levels in serum and vitreous humor varies according to the condition of the BRB. Therefore, in addition to measuring the serum NAb level, physicians should examine BRB integrity when planning retinal gene therapy. Translational Relevance This study provides substantial basis for retinal gene therapy using AAVs and how maintenance of BRB integrity in target diseases should be considered.
Collapse
Affiliation(s)
- Suhwan Lee
- Department of Ophthalmology, Kangwon National University Hospital, Kangwon National University Graduate School of Medicine, Chuncheon, Korea
| | - Im Kyeung Kang
- Department of Microbiology, University of Ulsan College of Medicine, Seoul, Korea.,Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Korea
| | - Ji Hyun Kim
- Department of Microbiology, University of Ulsan College of Medicine, Seoul, Korea.,Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Korea
| | - Bok Kyoung Jung
- Department of Ophthalmology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Keerang Park
- Biopharmaceutical R&D Center, Chungbuk Health & Science University, Cheongju-si, Chungbuk, Korea
| | | | - Joo Yong Lee
- Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Korea.,Department of Ophthalmology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Heuiran Lee
- Department of Microbiology, University of Ulsan College of Medicine, Seoul, Korea.,Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
8
|
Adeno-Associated Viral Vector-Mediated mTOR Inhibition by Short Hairpin RNA Suppresses Laser-Induced Choroidal Neovascularization. MOLECULAR THERAPY-NUCLEIC ACIDS 2017; 8:26-35. [PMID: 28918027 PMCID: PMC5477068 DOI: 10.1016/j.omtn.2017.05.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 05/29/2017] [Accepted: 05/29/2017] [Indexed: 12/20/2022]
Abstract
Choroidal neovascularization (CNV) is the defining characteristic feature of the wet subtype of age-related macular degeneration (AMD) and may result in irreversible blindness. Based on anti-vascular endothelial growth factor (anti-VEGF), the current therapeutic approaches to CNV are fraught with difficulties, and mammalian target of rapamycin (mTOR) has recently been proposed as a possible therapeutic target, although few studies have been conducted. Here, we show that a recombinant adeno-associated virus-delivered mTOR-inhibiting short hairpin RNA (rAAV-mTOR shRNA), which blocks the activity of both mTOR complex 1 and 2, represents a promising therapeutic approach for the treatment of CNV. Eight-week-old male C57/B6 mice were treated with the short hairpin RNA (shRNA) after generating CNV lesions in the eyes via laser photocoagulation. The recombinant adeno-associated virus (rAAV) delivery vehicle was able to effectively transduce cells in the inner retina, and significantly fewer inflammatory cells and less extensive CNV were observed in the animals treated with rAAV-mTOR shRNA when compared with control- and rAAV-scrambled shRNA-treated groups. Presumably related to the reduction of CNV, increased autophagy was detected in CNV lesions treated with rAAV-mTOR shRNA, whereas significantly fewer apoptotic cells detected in the outer nuclear layer around the CNV indicate that mTOR inhibition may also have neuroprotective effects. Taken together, these results demonstrate the therapeutic potential of mTOR inhibition, resulting from rAAV-mTOR shRNA activity, in the treatment of AMD-related CNV.
Collapse
|
9
|
Woo HN, Lee WI, Kim JH, Ahn J, Han JH, Lim SY, Lee WW, Lee H. Combined antitumor gene therapy with herpes simplex virus-thymidine kinase and short hairpin RNA specific for mammalian target of rapamycin. Int J Oncol 2015; 47:2233-9. [PMID: 26459571 DOI: 10.3892/ijo.2015.3194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/25/2015] [Indexed: 11/06/2022] Open
Abstract
A proof-of-concept study is presented using dual gene therapy that employed a small hairpin RNA (shRNA) specific for mammalian target of rapamycin (mTOR) and a herpes simplex virus-thymidine kinase (HSV-TK) gene to inhibit the growth of tumors. Recombinant adeno-associated virus (rAAV) vectors containing a mutant TK gene (sc39TK) were transduced into HeLa cells, and the prodrug ganciclovir (GCV) was administered to establish a suicide gene-therapy strategy. Additionally, rAAV vectors expressing an mTOR-targeted shRNA were employed to suppress mTOR-dependent tumor growth. GCV selectively induced death in tumor cells expressing TK, and the mTOR-targeted shRNA altered the cell cycle to impair tumor growth. Combining the TK-GCV system with mTOR inhibition suppressed tumor growth to a greater extent than that achieved with either treatment alone. Furthermore, HSV-TK expression and mTOR inhibition did not mutually interfere with each other. In conclusion, gene therapy that combines the TK-GCV system and mTOR inhibition shows promise as a novel strategy for cancer therapy.
Collapse
Affiliation(s)
- Ha-Na Woo
- Department of Microbiology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Won Il Lee
- Department of Microbiology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ji Hyun Kim
- Department of Microbiology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeonghyun Ahn
- Department of Microbiology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeong Hee Han
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sue Yeon Lim
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Won Woo Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Heuiran Lee
- Department of Microbiology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
10
|
Guo B, Zhang B, Zheng L, Tang T, Liu J, Wu H, Yang Z, Peng S, He X, Zhang H, Yue KKM, He F, Zhang L, Qin L, Bian Z, Tan W, Liang Z, Lu A, Zhang G. Therapeutic RNA interference targeting CKIP-1 with a cross-species sequence to stimulate bone formation. Bone 2014; 59:76-88. [PMID: 24246247 DOI: 10.1016/j.bone.2013.11.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 11/06/2013] [Accepted: 11/10/2013] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Casein kinase 2 interacting protein 1 (CKIP-1) is a newly discovered intracellular negative regulator of bone formation without affecting bone resorption. In this study, we aimed to identify a cross-species siRNA sequence targeting CKIP-1 to facilitate developing a novel RNAi-based bone anabolic drug for reversing established osteoporosis. METHODS Eight specifically designed cross-species CKIP-1 siRNA sequences were screened in human, rhesus, rat and mouse osteoblast-like cells in vitro to identify the optimal sequence with the highest knockdown efficiency. The effect of this optimal siRNA sequence on osteogenic differentiation and matrix mineralization was further examined in osteoblast-like cells across different species, followed by an immunogenicity assessment in human peripheral blood mononuclear cells in vitro. The intra-osseous localization and silencing efficiency of the optimal siRNA were examined in vivo using a biophotonic system and real-time polymerase chain reaction, respectively. The RNAi-mediated cleavage of the CKIP-1 transcript was confirmed by rapid amplification of the 5' cDNA ends in vivo. Furthermore, the effect of the optimal siRNA sequence on osteogenic differentiation, bone turnover biomarkers, bone mass and micro-architecture parameters was investigated in healthy and osteoporotic rodents. RESULTS The CKIP-1 siRNA sequence (si-3) was identified as the optimal sequence, which consistently maintained CKIP-1 mRNA/protein expression at the lowest level across species in vitro. The si-3 significantly increased mRNA expression levels of osteoblast phenotypic genes and matrix mineralization across species without inducing an immunostimulatory activity in vitro. The intra-osseous localization and RNAi-mediated CKIP-1 silencing with high efficiency were confirmed in vivo. Periodic intravenous injections of si-3 promoted mRNA expression of osteoblast phenotypic genes, enhanced bone formation, increased bone mass and elevated serum level of bone formation marker without raising urine level of bone resorption marker in the healthy rodents. Moreover, the si-3 treatment promoted bone formation, improved trabecular micro-architecture and reversed bone loss in the osteoporotic mice. CONCLUSIONS The identified optimal CKIP-1 siRNA sequence (si-3) could promote osteogenic differentiation across species in vitro, stimulate bone formation in the healthy rodents and reverse bone loss in the osteoporotic mice.
Collapse
Affiliation(s)
- Baosheng Guo
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Hong Kong Baptist University Branch of State Key Laboratory of Chemo/Biosensing and Chemometrics of Hunan University, Hong Kong, China; Institute of Integrated Bioinfomedicine & Translational Science, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China; Academician CHAN Sun Chi Albert Workroom for Advancing Translational Medicine in Bone & Joint Diseases, Kunshan RNAi Institute, Kunshan Industrial Technology Research Institute, Kunshan, Jiangsu, China
| | - Baoting Zhang
- School of Chinese Medicine, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, China.
| | - Lizhen Zheng
- School of Chinese Medicine, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Tao Tang
- School of Chinese Medicine, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Jin Liu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Hong Kong Baptist University Branch of State Key Laboratory of Chemo/Biosensing and Chemometrics of Hunan University, Hong Kong, China; Institute of Integrated Bioinfomedicine & Translational Science, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China; Academician CHAN Sun Chi Albert Workroom for Advancing Translational Medicine in Bone & Joint Diseases, Kunshan RNAi Institute, Kunshan Industrial Technology Research Institute, Kunshan, Jiangsu, China
| | - Heng Wu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Hong Kong Baptist University Branch of State Key Laboratory of Chemo/Biosensing and Chemometrics of Hunan University, Hong Kong, China; Institute of Integrated Bioinfomedicine & Translational Science, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China; Academician CHAN Sun Chi Albert Workroom for Advancing Translational Medicine in Bone & Joint Diseases, Kunshan RNAi Institute, Kunshan Industrial Technology Research Institute, Kunshan, Jiangsu, China
| | - Zhijun Yang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Hong Kong Baptist University Branch of State Key Laboratory of Chemo/Biosensing and Chemometrics of Hunan University, Hong Kong, China; Institute of Integrated Bioinfomedicine & Translational Science, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China; Academician CHAN Sun Chi Albert Workroom for Advancing Translational Medicine in Bone & Joint Diseases, Kunshan RNAi Institute, Kunshan Industrial Technology Research Institute, Kunshan, Jiangsu, China
| | - Songlin Peng
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Hong Kong Baptist University Branch of State Key Laboratory of Chemo/Biosensing and Chemometrics of Hunan University, Hong Kong, China; Institute of Integrated Bioinfomedicine & Translational Science, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Xiaojuan He
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Hongqi Zhang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Hong Kong Baptist University Branch of State Key Laboratory of Chemo/Biosensing and Chemometrics of Hunan University, Hong Kong, China; Institute of Integrated Bioinfomedicine & Translational Science, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Kevin K M Yue
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Hong Kong Baptist University Branch of State Key Laboratory of Chemo/Biosensing and Chemometrics of Hunan University, Hong Kong, China; Institute of Integrated Bioinfomedicine & Translational Science, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Fuchu He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Ling Qin
- School of Chinese Medicine, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhaoxiang Bian
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Hong Kong Baptist University Branch of State Key Laboratory of Chemo/Biosensing and Chemometrics of Hunan University, Hong Kong, China; Institute of Integrated Bioinfomedicine & Translational Science, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China; Academician CHAN Sun Chi Albert Workroom for Advancing Translational Medicine in Bone & Joint Diseases, Kunshan RNAi Institute, Kunshan Industrial Technology Research Institute, Kunshan, Jiangsu, China
| | - Weihong Tan
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Hong Kong Baptist University Branch of State Key Laboratory of Chemo/Biosensing and Chemometrics of Hunan University, Hong Kong, China; Institute of Integrated Bioinfomedicine & Translational Science, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China; Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha, China
| | - Zicai Liang
- Academician CHAN Sun Chi Albert Workroom for Advancing Translational Medicine in Bone & Joint Diseases, Kunshan RNAi Institute, Kunshan Industrial Technology Research Institute, Kunshan, Jiangsu, China; Laboratory of Nucleic Acid Technology, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Aiping Lu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Hong Kong Baptist University Branch of State Key Laboratory of Chemo/Biosensing and Chemometrics of Hunan University, Hong Kong, China; Institute of Integrated Bioinfomedicine & Translational Science, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China; Academician CHAN Sun Chi Albert Workroom for Advancing Translational Medicine in Bone & Joint Diseases, Kunshan RNAi Institute, Kunshan Industrial Technology Research Institute, Kunshan, Jiangsu, China; Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Ge Zhang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Hong Kong Baptist University Branch of State Key Laboratory of Chemo/Biosensing and Chemometrics of Hunan University, Hong Kong, China; Institute of Integrated Bioinfomedicine & Translational Science, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China; Academician CHAN Sun Chi Albert Workroom for Advancing Translational Medicine in Bone & Joint Diseases, Kunshan RNAi Institute, Kunshan Industrial Technology Research Institute, Kunshan, Jiangsu, China.
| |
Collapse
|