1
|
Gong X, Gao H, Wang W, Xu T. Intramuscular Injection of rAAV2-retro for Low Motor Neuron Transduction: Evaluating Five Promoters. Int J Med Sci 2025; 22:775-789. [PMID: 39991760 PMCID: PMC11843134 DOI: 10.7150/ijms.101807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 12/12/2024] [Indexed: 02/25/2025] Open
Abstract
Recombinant adeno-associated viral vectors (rAAVs) can effectively deliver transgene to the nervous system. The selection of AAV serotype and promoter significantly influences the dynamics of the transgene expression, including its strength and cell-specificity. Previous studies demonstrated that in neonatal mice, the intramuscular (IM) injection of the rAAV2-retro vector could efficiently deliver transgene to lower motor neurons (LMNs) of the brainstem and spinal cord. However, the best promoter for the expression of transgene in the central neural system (CNS) using rAAV2-retro remains undetermined. This study compared five commonly used promoters, including mouse phosphoglycerate kinase (mPGK), CMV early enhancer/chicken β-actin/short β-globulin intron (CAG), human cytomegalovirus (hCMV), chicken β-actin (CBA), and human synapsin (hSyn) promoters. The IM (unilateral gastrocnemius muscle) injection of rAAV2-retro vectors packaged with the reporter constructs containing each promoter was performed in the newborn C57BL/6J mice. The levels of gene expression and the types of cells were examined using the light-sheet illumination imaging technique and confocal microscopy. Our findings revealed that rAAV2-retro primarily targeted the brainstem and spinal cord within the CNS. Among the five promoters tested, CAG and hCMV showed the highest gene expression. Almost all the transduced cells were identified as LMNs. Additionally, gene expression driven by hCMV was found to be dependent of the inclusion of WPRE and β-globin intron elements. Importantly, none of the promoters induced hepatotoxicity, ensuring the safety of rAAV2-retro-mediated expression. This study provided valuable insights for optimizing the rAAV2-retro-mediated gene delivery system to LMNs in the brainstem and spinal cord, which might have potential implications for research on motor neuron-related diseases.
Collapse
Affiliation(s)
- Xueqi Gong
- Laboratory Animal Center, Fudan University, Shanghai 200032, China
- Laboratory Animal Resource Center, Fudan University, Shanghai 200032, China
| | - Haitong Gao
- Laboratory Animal Center, Fudan University, Shanghai 200032, China
- Laboratory Animal Resource Center, Fudan University, Shanghai 200032, China
| | - Wenyuan Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese academy of Science, Shanghai 200032, China
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Tonghui Xu
- Laboratory Animal Center, Fudan University, Shanghai 200032, China
- Laboratory Animal Resource Center, Fudan University, Shanghai 200032, China
| |
Collapse
|
2
|
Thakore P, Karki S, Hrdlicka HC, Garcia-Munoz J, Pereira RC, Delany AM. Decreasing miR-433-3p Activity in the Osteoblast Lineage Blunts Glucocorticoid-mediated Bone Loss. Endocrinology 2025; 166:bqaf008. [PMID: 39820728 PMCID: PMC11791524 DOI: 10.1210/endocr/bqaf008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/05/2024] [Accepted: 01/15/2025] [Indexed: 01/19/2025]
Abstract
Glucocorticoid excess causes bone loss due to decreased bone formation and increased bone resorption; miR-433-3p is a microRNA (miRNA) that negatively regulates bone formation in male mice by targeting Runx2 as well as RNAs involved in Wnt, protein kinase A, and endogenous glucocorticoid signaling. To examine the impact of miR-433-3p on glucocorticoid-mediated bone loss, transgenic mice expressing a miR-433-3p tough decoy inhibitor in the osteoblast lineage were administered prednisolone via slow-release pellets. Bone loss was greater in control mice treated with prednisolone compared with miR-433-3p tough decoy mice due to higher osteoclast activity in the controls. In whole femurs, Rankl was significantly higher in prednisolone-treated controls compared with miR-433-3p tough decoy mice. Surprisingly, negative regulators of Wnt signaling Sost and Dkk1 were higher in miR-433-3p tough decoy mice and were unaffected by prednisolone. Luciferase- 3'-untranslated region reporter assays demonstrated that Sost is a novel miR-433-3p target, whereas Dkk1 is a previously validated miR-433-3p target. miR-433-3p levels are lower in matrix-synthesizing osteoblasts than in more osteocytic cells; thus the impact of miR-433-3p on the osteoblast lineage may be dependent on cell context: it is a negative regulator in matrix-depositing osteoblasts by targeting RNAs important for differentiation and function but a positive regulator in osteocytes, due to its ability to target prominently expressed negative regulators of Wnt signaling, Sost and Dkk1. The mechanisms by which miR-433-3p indirectly regulates glucocorticoid-mediated osteoclastogenesis remain unknown. However, we speculate that this regulation may be mediated by miR-433-3p activity in osteocytes, which play an important role in controlling osteoclastogenesis.
Collapse
Affiliation(s)
- Prachi Thakore
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| | - Sangita Karki
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| | - Henry C Hrdlicka
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| | - John Garcia-Munoz
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| | - Renata C Pereira
- Division of Pediatric Nephrology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Anne M Delany
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| |
Collapse
|
3
|
Duncan CJ, Skouboe MK, Howarth S, Hollensen AK, Chen R, Børresen ML, Thompson BJ, Stremenova Spegarova J, Hatton CF, Stæger FF, Andersen MK, Whittaker J, Paludan SR, Jørgensen SE, Thomsen MK, Mikkelsen JG, Heilmann C, Buhas D, Øbro NF, Bay JT, Marquart HV, de la Morena MT, Klejka JA, Hirschfeld M, Borgwardt L, Forss I, Masmas T, Poulsen A, Noya F, Rouleau G, Hansen T, Zhou S, Albrechtsen A, Alizadehfar R, Allenspach EJ, Hambleton S, Mogensen TH. Life-threatening viral disease in a novel form of autosomal recessive IFNAR2 deficiency in the Arctic. J Exp Med 2022; 219:e20212427. [PMID: 35442417 PMCID: PMC9026249 DOI: 10.1084/jem.20212427] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 02/28/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022] Open
Abstract
Type I interferons (IFN-I) play a critical role in human antiviral immunity, as demonstrated by the exceptionally rare deleterious variants of IFNAR1 or IFNAR2. We investigated five children from Greenland, Canada, and Alaska presenting with viral diseases, including life-threatening COVID-19 or influenza, in addition to meningoencephalitis and/or hemophagocytic lymphohistiocytosis following live-attenuated viral vaccination. The affected individuals bore the same homozygous IFNAR2 c.157T>C, p.Ser53Pro missense variant. Although absent from reference databases, p.Ser53Pro occurred with a minor allele frequency of 0.034 in their Inuit ancestry. The serine to proline substitution prevented cell surface expression of IFNAR2 protein, small amounts of which persisted intracellularly in an aberrantly glycosylated state. Cells exclusively expressing the p.Ser53Pro variant lacked responses to recombinant IFN-I and displayed heightened vulnerability to multiple viruses in vitro-a phenotype rescued by wild-type IFNAR2 complementation. This novel form of autosomal recessive IFNAR2 deficiency reinforces the essential role of IFN-I in viral immunity. Further studies are warranted to assess the need for population screening.
Collapse
Affiliation(s)
- Christopher J.A. Duncan
- Clinical and Translational Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Morten K. Skouboe
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Sophie Howarth
- Clinical and Translational Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Anne K. Hollensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Rui Chen
- Clinical and Translational Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Malene L. Børresen
- Department of Paediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Benjamin J. Thompson
- Clinical and Translational Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Jarmila Stremenova Spegarova
- Clinical and Translational Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Catherine F. Hatton
- Clinical and Translational Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Frederik F. Stæger
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Mette K. Andersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - John Whittaker
- Clinical and Translational Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
| | | | - Sofie E. Jørgensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | | | | | - Carsten Heilmann
- Department of Paediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Medical Department, Pediatric Section, Dronning Ingrid Hospital, Nuuk, Greenland
| | - Daniela Buhas
- Division of Genetics, Department of Specialized Medicine, McGill University Health Centre, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Nina F. Øbro
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Jakob T. Bay
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Hanne V. Marquart
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | - M. Teresa de la Morena
- Seattle Children’s Hospital, Seattle, WA
- Department of Pediatrics, University of Washington, Seattle, WA
| | | | | | - Line Borgwardt
- Center for Genomic Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Isabel Forss
- Center for Genomic Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Tania Masmas
- Department of Paediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Anja Poulsen
- Department of Paediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Francisco Noya
- Division of Allergy & Clinical Immunology, Montreal Children’s Hospital, Montreal General Hospital, McGill University, Montreal, Quebec, Canada
| | - Guy Rouleau
- The Neuro, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sirui Zhou
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Anders Albrechtsen
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Reza Alizadehfar
- Division of Allergy & Clinical Immunology, Montreal Children’s Hospital, Montreal General Hospital, McGill University, Montreal, Quebec, Canada
| | - Eric J. Allenspach
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
- Seattle Children’s Hospital, Seattle, WA
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA
- Brotman Baty Institute for Precision Medicine, Seattle, WA
| | - Sophie Hambleton
- Clinical and Translational Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne, UK
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Trine H. Mogensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
4
|
Guo Z, Xie M, Zou Y, Liang Q, Liu F, Su J, He Z, Cai X, Chen Z, Zhao Q, Zhao K. Circular RNA Hsa_circ_0006766 targets microRNA miR-4739 to regulate osteogenic differentiation of human bone marrow mesenchymal stem cells. Bioengineered 2021; 12:5679-5687. [PMID: 34524066 PMCID: PMC8806466 DOI: 10.1080/21655979.2021.1967712] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 08/10/2021] [Indexed: 01/10/2023] Open
Abstract
Circular RNAs (circRNAs) are emerging as important regulators in bone metabolism, which is mediated by microRNA (miRNA) sponges. However, it is not clear how circRNA regulates osteogenic differentiation of human bone marrow mesenchymal stem cells (hBM-MSCs).Therefore, based on the previous circRNA chip results, hsa_circ_0006766, which is differentially expressed in the osteogenic differentiation of hBM-MSCs, was screened out, and bioinformatics analysis was performed to predict potential target miRNAs. During osteogenic differentiation of hBM-MSCs, hsa_circ_0006766 and its target miRNAs (miR-4739, miR-619-5p, miR-5787, miR-7851-3p, and miR-3192-5p) were detected by quantitative Real Time-PCR (qRT-PCR). Target gene prediction for the differentially expressed target miRNAs was performed, and target genes were validated by dual-luciferase reporter gene assay and qRT-PCR. It is shown that hsa_circ_0006766 was up-regulated and miR-4739 was down-regulated during osteogenic differentiation of hBM-MSCs.Moreover, the target gene Notch2 was predicted to be highly expressed during osteogenic differentiation. And dual-luciferase assay proved that Notch2 was the gene targeting to miR-4739. Taken together, our finding confirmed that hsa_circ_0006766 may act as a major regulatory part in osteogenic differentiation of hBM-MSCs via an hsa_circ_0006766-miR-4739-Notch2 regulatory axis. Accordingly, hsa_circ_0006766 may affect the development of osteoporosis and may thus become a therapeutic target.
Collapse
Affiliation(s)
- Zhaodi Guo
- The Clinical laboratory, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Manlin Xie
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yanfang Zou
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Qianxin Liang
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Fubin Liu
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jing Su
- The Clinical laboratory, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhiliang He
- The Clinical laboratory, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiuping Cai
- The Clinical laboratory, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhixiang Chen
- The Clinical laboratory, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Qing Zhao
- The Clinical laboratory, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Kewei Zhao
- The Clinical laboratory, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Zhang J, Li D, Zhang R, Peng R, Li J. Delivery of microRNA-21-sponge and pre-microRNA-122 by MS2 virus-like particles to therapeutically target hepatocellular carcinoma cells. Exp Biol Med (Maywood) 2021; 246:2463-2472. [PMID: 34644206 DOI: 10.1177/15353702211035689] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
MicroRNAs are related to the development of hepatocellular carcinoma and can serve as potential therapeutic targets. Therapeutic strategies increasing tumor-suppressive microRNAs and reducing oncogenic microRNAs have been developed. Herein, the effects of simultaneously altering two microRNAs using MS2 virus-like particles were studied. The sequences of microRNA-21-sponge and pre-microRNA-122 were connected and cloned into a virus-like particle expression vector. Virus-like particles containing microRNA-21-sponge and pre-microRNA-122 sequences were prepared and crosslinked with a cell-specific peptide targeting hepatocellular carcinoma cells. Delivery effects were studied using RT-qPCR and functional assays to investigate the level of target mRNAs, cell toxicity, and the effects of proliferation, invasion, and migration. Virus-like particles delivered miR-21-sponge into cells, with the Ct value reaching 10 at most. The linked pre-miR-122 was processed into mature miR-122. The mRNA targets of miR-21 were derepressed as predicted and upregulated 1.2-2.8-fold, and the expression of proteins was elevated correspondingly. Proliferation, migration, and invasion of HCC cells were inhibited by miR-21-sponge. Simultaneous delivery of miR-21-sponge and miR-122 further decreased proliferation, migration, and invasion by up to 34%, 63%, and 65%, respectively. And the combination promoted the apoptosis of HCC cells. In conclusion, delivering miR-21-sponge and miR-122 using virus-like particles modified by cell-specific peptides is an effective and convenient strategy to correct microRNA dysregulation in hepatocellular carcinoma cells and is a promising therapeutic strategy for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Jiawei Zhang
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China.,Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing 100730, P.R. China
| | - Dandan Li
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China.,Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing 100730, P.R. China
| | - Rui Zhang
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China.,Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing 100730, P.R. China
| | - Rongxue Peng
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China.,Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing 100730, P.R. China
| | - Jinming Li
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China.,Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing 100730, P.R. China
| |
Collapse
|
6
|
Shin B, Hrdlicka HC, Delany AM, Lee SK. Inhibition of miR-29 Activity in the Myeloid Lineage Increases Response to Calcitonin and Trabecular Bone Volume in Mice. Endocrinology 2021; 162:bqab135. [PMID: 34192317 PMCID: PMC8328098 DOI: 10.1210/endocr/bqab135] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Indexed: 12/29/2022]
Abstract
The miR-29-3p family (miR-29a, miR-29b, miR-29c) of microRNAs is increased during receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclastogenesis. In vivo, activation of a miR-29-3p tough decoy inhibitor in Cre recombinase under the control of the lysozyme 2 promoter-expressing cells (myeloid lineage) resulted in mice displaying enhanced trabecular and cortical bone volume because of decreased bone resorption. Calcitonin receptor (Calcr) is a miR-29 target that negatively regulates bone resorption. CALCR was significantly increased in RANKL-treated miR-29-decoy osteoclasts, and these cells were more responsive to the inhibitory effect of calcitonin on osteoclast formation. Further, cathepsin K (Ctsk), which is critical for resorption, was decreased in miR-29-decoy cells. CALCR is a Gs-coupled receptor and its activation raises cAMP levels. In turn, cAMP suppresses cathepsin K, and cAMP levels were increased in miR-29-decoy cells. siRNA-mediated knock-down of Calcr in miR-29 decoy osteoclasts allowed recovery of cathepsin K levels in these cells. Overall, using a novel knockin tough decoy mouse model, we identified a new role for miR-29-3p in bone homeostasis. In RANKL-driven osteoclastogenesis, as seen in normal bone remodeling, miR-29-3p promotes resorption. Consequently, inhibition of miR-29-3p activity in the myeloid lineage leads to increased trabecular and cortical bone. Further, this study documents an interrelationship between CALCR and CTSK in osteoclastic bone resorption, which is modulated by miR-29-3p.
Collapse
Affiliation(s)
- Bongjin Shin
- Center on Aging, UConn Health, Farmington, CT 06030, USA
- Department of Biological Sciences, University at Buffalo, Buffalo, NY 14260, USA
| | - Henry C Hrdlicka
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| | - Anne M Delany
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| | - Sun-Kyeong Lee
- Center on Aging, UConn Health, Farmington, CT 06030, USA
| |
Collapse
|
7
|
Comparison of the expression and toxicity of AAV2/9 carrying the human A53T α-synuclein gene in presence or absence of WPRE. Heliyon 2021; 7:e06302. [PMID: 33665452 PMCID: PMC7903312 DOI: 10.1016/j.heliyon.2021.e06302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/25/2020] [Accepted: 02/12/2021] [Indexed: 12/15/2022] Open
Abstract
Woodchuck Hepatitis Virus Post-transcriptional Regulatory Element (WPRE) is thought to enhance transgene expression of target genes delivered by adeno-associated viral (AAV) vectors. This study assessed the protein expression of α-synuclein, phosphorylated α-synuclein at Serine 129, extent of nigrostriatal degeneration as well as subsequent behavioral deficits induced by unilateral intranigral stereotactic injection in male adult C57BL/6J mice of an AAV2/9 expressing A53T human α-synuclein under the control of the synapsin promoter in presence or absence of the WPRE. The presence of WPRE enabled to achieve greater nigrostriatal degeneration and synucleinopathy which was concomitant with worsened forelimb use asymmetry. This work refines a mouse Parkinson's disease model in which anatomo-pathology is related to behavioral deficits.
Collapse
|
8
|
Hollensen AK, Thomsen HS, Lloret-Llinares M, Kamstrup AB, Jensen JM, Luckmann M, Birkmose N, Palmfeldt J, Jensen TH, Hansen TB, Damgaard CK. circZNF827 nucleates a transcription inhibitory complex to balance neuronal differentiation. eLife 2020; 9:e58478. [PMID: 33174841 PMCID: PMC7657652 DOI: 10.7554/elife.58478] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 10/29/2020] [Indexed: 12/20/2022] Open
Abstract
Circular RNAs are important for many cellular processes but their mechanisms of action remain poorly understood. Here, we map circRNA inventories of mouse embryonic stem cells, neuronal progenitor cells and differentiated neurons and identify hundreds of highly expressed circRNAs. By screening several candidate circRNAs for a potential function in neuronal differentiation, we find that circZNF827 represses expression of key neuronal markers, suggesting that this molecule negatively regulates neuronal differentiation. Among 760 tested genes linked to known neuronal pathways, knockdown of circZNF827 deregulates expression of numerous genes including nerve growth factor receptor (NGFR), which becomes transcriptionally upregulated to enhance NGF signaling. We identify a circZNF827-nucleated transcription-repressive complex containing hnRNP-K/L proteins and show that knockdown of these factors strongly augments NGFR regulation. Finally, we show that the ZNF827 protein is part of the mRNP complex, suggesting a functional co-evolution of a circRNA and the protein encoded by its linear pre-mRNA host.
Collapse
Affiliation(s)
| | | | - Marta Lloret-Llinares
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
- European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Wellcome Genome Campus, HinxtonCambridgeUnited Kingdom
| | | | | | - Majbritt Luckmann
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| | - Nanna Birkmose
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| | - Johan Palmfeldt
- Department of Clinical Medicine, Research Unit for Molecular Medicine, Aarhus UniversityAarhusDenmark
| | - Torben Heick Jensen
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| | - Thomas B Hansen
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| | | |
Collapse
|
9
|
Due H, Schönherz AA, Ryø L, Primo MN, Jespersen DS, Thomsen EA, Roug AS, Xiao M, Tan X, Pang Y, Young KH, Bøgsted M, Mikkelsen JG, Dybkær K. MicroRNA-155 controls vincristine sensitivity and predicts superior clinical outcome in diffuse large B-cell lymphoma. Blood Adv 2019; 3:1185-1196. [PMID: 30967394 PMCID: PMC6457225 DOI: 10.1182/bloodadvances.2018029660] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/04/2019] [Indexed: 12/17/2022] Open
Abstract
A major clinical challenge of diffuse large B-cell lymphoma (DLBCL) is that up to 40% of patients have refractory disease or relapse after initial response to therapy as a result of drug-specific molecular resistance. The purpose of the present study was to investigate microRNA (miRNA) involvement in vincristine resistance in DLBCL, which was pursued by functional in vitro analysis in DLBCL cell lines and by outcome analysis of patients with DLBCL treated with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP). Differential miRNA expression analysis identified miR-155 as highly expressed in vincristine-sensitive DLBCL cell lines compared with resistant ones. Ectopic upregulation of miR-155 sensitized germinal-center B-cell-like (GCB)-DLBCL cell lines to vincristine, and consistently, reduction and knockout of miR-155 induced vincristine resistance, documenting that miR-155 functionally induces vincristine sensitivity. Target gene analysis identified miR-155 as inversely correlated with Wee1, supporting Wee1 as a target of miR-155 in DLBCL. Chemical inhibition of Wee1 sensitized GCB cells to vincristine, suggesting that miR-155 controls vincristine response through Wee1. Outcome analysis in clinical cohorts of DLBCL revealed that high miR-155 expression level was significantly associated with superior survival for R-CHOP-treated patients of the GCB subclass, independent of international prognostic index, challenging the commonly accepted perception of miR-155 as an oncomiR. However, miR-155 did not provide prognostic information when analyzing the entire DLBCL cohort or activated B-cell-like classified patients. In conclusion, we experimentally confirmed a direct link between high miR-155 expression and vincristine sensitivity in DLBCL and documented an improved clinical outcome of GCB-classified patients with high miR-155 expression level.
Collapse
Affiliation(s)
- Hanne Due
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark
| | - Anna Amanda Schönherz
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, and
| | - Laura Ryø
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | | | | | | | - Min Xiao
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xiaohong Tan
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yuyang Pang
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ken H Young
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Martin Bøgsted
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark
- Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; and
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | | - Karen Dybkær
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark
- Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; and
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
10
|
Hooykaas MJG, Soppe JA, De Buhr HM, Kruse E, Wiertz EJHJ, Lebbink RJ. RNA accessibility impacts potency of Tough Decoy microRNA inhibitors. RNA Biol 2018; 15:1410-1419. [PMID: 30339041 PMCID: PMC6284568 DOI: 10.1080/15476286.2018.1537746] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
MicroRNAs (miRNAs) are small RNA molecules that post-transcriptionally regulate gene expression through silencing of complementary target mRNAs. miRNAs are involved in many biological processes, including cell proliferation, differentiation, cell signaling and cellular defense responses to infection. Strategies that allow for strong and stable suppression of specific microRNA activity are needed to study miRNA functions and to develop therapeutic intervention strategies aimed at interfering with miRNA activity in vivo. One of these classes of miRNA inhibitors are Tough Decoys (TuD) RNAs, which comprise of an imperfect RNA hairpin structure that harbors two opposing miRNA binding sites. Upon developing TuDs targeting Epstein-Barr virus miRNAs, we observed a strong variation in inhibitory potential between different TuD RNAs targeting the same miRNA. We show that the composition of the 'bulge' sequence in the miRNA binding sites has a strong impact on the inhibitory potency of the TuD. Our data implies that miRNA inhibition correlates with the thermodynamic properties of the TuD and that design aimed at lowering the TuD opening energy increases TuD potency. Our study provides specific guidelines for the design and construction of potent decoy-based miRNA inhibitors, which may be used for future therapeutic intervention strategies.
Collapse
Affiliation(s)
- Marjolein J G Hooykaas
- a Department of Medical Microbiology , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Jasper A Soppe
- a Department of Medical Microbiology , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Hendrik M De Buhr
- a Department of Medical Microbiology , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Elisabeth Kruse
- a Department of Medical Microbiology , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Emmanuel J H J Wiertz
- a Department of Medical Microbiology , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Robert J Lebbink
- a Department of Medical Microbiology , University Medical Center Utrecht , Utrecht , The Netherlands
| |
Collapse
|
11
|
Enhanced Tailored MicroRNA Sponge Activity of RNA Pol II-Transcribed TuD Hairpins Relative to Ectopically Expressed ciRS7-Derived circRNAs. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 13:365-375. [PMID: 30347350 PMCID: PMC6198105 DOI: 10.1016/j.omtn.2018.09.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 09/17/2018] [Accepted: 09/17/2018] [Indexed: 12/30/2022]
Abstract
As key regulators of gene expression, microRNAs (miRNAs) have emerged as targets in basic experimentation and therapy. Administration of DNA-encoded RNA molecules, targeting miRNAs through base pairing, is one viable strategy for inhibiting specific miRNAs. A naturally occurring circular RNA (circRNA), ciRS-7, serving as a miRNA-7 (miR-7) sponge was recently identified. This has sparked tremendous interest in adapting circRNAs for suppressing miRNA function. In parallel, we and others have demonstrated efficacy of expressed anti-miRNA Tough Decoy (TuD) hairpins. To compare properties of such inhibitors, we express ciRS-7 and TuD-containing miRNA suppressor transcripts from identical vector formats adapted from RNA polymerase II-directed expression plasmids previously used for production of ciRS-7. In general, markedly higher levels of miR-7 suppression with TuD transcripts relative to ciRS-7 are observed, leading to superior miRNA sponge effects using expressed TuD hairpins. Notably however, we find that individual ciRS-7 transcripts are more potent inhibitors of miR-7 activity than individual TuD7-containing transcripts, although each miR-7 seed match target site in ciRS-7 is, on average, less potent than the perfectly matched target sites in the TuD motif. All together, our studies call for improved means of designing and producing circRNAs for customized miRNA targeting to match TuD hairpins for tailored miRNA suppression.
Collapse
|
12
|
Bernardo BC, Gregorevic P, Ritchie RH, McMullen JR. Generation of MicroRNA-34 Sponges and Tough Decoys for the Heart: Developments and Challenges. Front Pharmacol 2018; 9:1090. [PMID: 30298011 PMCID: PMC6160554 DOI: 10.3389/fphar.2018.01090] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/07/2018] [Indexed: 12/13/2022] Open
Abstract
Heart failure (HF) is a debilitating and deadly chronic disease, with almost 50% of patients with HF dying within 5 years of diagnosis. With limited effective therapies to treat or cure HF, new therapies are greatly needed. microRNAs (miRNAs) are small non-coding RNA molecules that are powerful regulators of gene expression and play a key role in almost every biological process. Disruptions in miRNA gene expression has been functionally linked to numerous diseases, including cardiovascular disease. Molecular tools for manipulating miRNA activity have been developed, and there is evidence from preclinical studies demonstrating the potential of miRNAs to be therapeutic targets for cardiovascular disease. For clinical application, miRNA sponges and tough decoys have been developed for more stable suppression and targeted delivery of the miRNA of choice. The aim of this study was to generate miRNA sponges and tough decoys to target miR-34 in the mouse heart. We present data to show that using both approaches we were unable to get significant knockdown of miR-34 or regulate miR-34 target genes in the heart in vivo. We also review recent applications of this method in the heart and discuss further considerations for optimisation in construct design and testing, and the obstacles to be overcome before they enter the clinic.
Collapse
Affiliation(s)
- Bianca C Bernardo
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia.,Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, Australia
| | - Paul Gregorevic
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Physiology, Centre for Muscle Research, The University of Melbourne, Melbourne, VIC, Australia
| | - Rebecca H Ritchie
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, Australia.,Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, VIC, Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, Australia.,Department of Medicine, Monash University, Clayton, VIC, Australia.,Department of Physiology, Monash University, Clayton, VIC, Australia.,Department of Physiology, Anatomy, and Microbiology, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|