601
|
Li X, Liang Q, Zhang W, Li Y, Ye J, Zhao F, Chen X, Wang S. Bio-inspired bioactive glasses for efficient microRNA and drug delivery. J Mater Chem B 2017; 5:6376-6384. [PMID: 32264454 DOI: 10.1039/c7tb01021d] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Bio-inspired pinecone-like bioactive glasses consisting of ordered thin-layers separated by consistent cavities were synthesized using a sol-gel process. The short diameter of the as-produced particles was as short as 161 nm, and the surface area was as high as 280 m2 g-1. The pore volume, ranging from ∼0.74 cm3 g-1 to ∼0.67 cm3 g-1, could be modulated by the aqueous ammonia concentration. The surface was further tailored for positive charges by amino grafting. The as-produced nanoparticles could successfully enter cells via endocytosis. The microRNA delivery of the bioactive glass particles was further investigated by fluorescence microscopy and flow cytometry, indicating a loading efficiency and transfection efficiency greater than 90%. The potential of such particles as drug carriers was also studied. CCK8, live-dead cell staining and PI/annexinV double staining analyses confirmed that the bioactive glass particles loaded with antitumour doxorubicin (DOX) significantly accelerated the apoptosis of tumour cells. These bio-inspired bioactive glasses are promising as novel vectors for drug and microRNA delivery with high efficiency.
Collapse
Affiliation(s)
- Xian Li
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, People's Republic of China.
| | | | | | | | | | | | | | | |
Collapse
|
602
|
Nabzdyk CS, Pradhan-Nabzdyk L, LoGerfo FW. RNAi therapy to the wall of arteries and veins: anatomical, physiologic, and pharmacological considerations. J Transl Med 2017; 15:164. [PMID: 28754174 PMCID: PMC5534068 DOI: 10.1186/s12967-017-1270-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 07/20/2017] [Indexed: 12/02/2022] Open
Abstract
Background Cardiovascular disease remains a major health care challenge. The knowledge about the underlying mechanisms of the respective vascular disease etiologies has greatly expanded over the last decades. This includes the contribution of microRNAs, endogenous non-coding RNA molecules, known to vastly influence gene expression. In addition, short interference RNA has been established as a mechanism to temporarily affect gene expression. This review discusses challenges relating to the design of a RNA interference therapy strategy for the modulation of vascular disease. Despite advances in medical and surgical therapies, atherosclerosis (ATH), aortic aneurysms (AA) are still associated with high morbidity and mortality. In addition, intimal hyperplasia (IH) remains a leading cause of late vein and prosthetic bypass graft failure. Pathomechanisms of all three entities include activation of endothelial cells (EC) and dedifferentiation of vascular smooth muscle cells (VSMC). RNA interference represents a promising technology that may be utilized to silence genes contributing to ATH, AA or IH. Successful RNAi delivery to the vessel wall faces multiple obstacles. These include the challenge of cell specific, targeted delivery of RNAi, anatomical barriers such as basal membrane, elastic laminae in arterial walls, multiple layers of VSMC, as well as adventitial tissues. Another major decision point is the route of delivery and potential methods of transfection. A plethora of transfection reagents and adjuncts have been described with varying efficacies and side effects. Timing and duration of RNAi therapy as well as target gene choice are further relevant aspects that need to be addressed in a temporo-spatial fashion. Conclusions While multiple preclinical studies reported encouraging results of RNAi delivery to the vascular wall, it remains to be seen if a single target can be sufficient to the achieve clinically desirable changes in the injured vascular wall in humans. It might be necessary to achieve simultaneous and/or sequential silencing of multiple, synergistically acting target genes. Some advances in cell specific RNAi delivery have been made, but a reliable vascular cell specific transfection strategy is still missing. Also, off-target effects of RNAi and unwanted effects of transfection agents on gene expression are challenges to be addressed. Close collaborative efforts between clinicians, geneticists, biologists, and chemical and medical engineers will be needed to provide tailored therapeutics for the various types of vascular diseases.
Collapse
Affiliation(s)
- Christoph S Nabzdyk
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Frank W. LoGerfo Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 110 Francis Street, Boston, MA, 02215, USA
| | - Leena Pradhan-Nabzdyk
- Frank W. LoGerfo Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 110 Francis Street, Boston, MA, 02215, USA.
| | - Frank W LoGerfo
- Frank W. LoGerfo Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 110 Francis Street, Boston, MA, 02215, USA
| |
Collapse
|
603
|
Diaz-Dussan D, Nakagawa Y, Peng YY, C LVS, Ebara M, Kumar P, Narain R. Effective and Specific Gene Silencing of Epidermal Growth Factor Receptors Mediated by Conjugated Oxaborole and Galactose-Based Polymers. ACS Macro Lett 2017; 6:768-774. [PMID: 35650860 DOI: 10.1021/acsmacrolett.7b00388] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Oxaborole-based polymers are stimuli-responsive materials that can reversibly interact with diols at pH values higher than their pKa. The strong binding of the oxaborole with cis-hydroxyl groups allow rapid cross-linking of the polymer chains. In this study, we exploited this phenomenon to develop a novel delivery system for the complexation, protection, and delivery of epidermal growth factor receptors (EGFR) siRNA (small interfering RNA). Galactose and oxaborole polymers were first synthesized by the reversible addition-fragmentation chain transfer (RAFT) process, and they were found to show a robust interaction with each other via the oxaborole-diol effect, which allowed the formation of stable polyplexes with siRNA. Although complexes were successfully formed between the neutral galactose and oxaborole-based polymers, these complexes were insufficient in the protection of the siRNA. Therefore, cationic glycopolymers and oxaborole polymers were investigated showing superior complexation with siRNA and exhibiting effective gene silencing in HeLa (cervical) cancer cells, while showing low toxicity. Gene silencing of up to 60% was achieved with these new complexes in the presence and absence of serum. The excellent stability of the complexes under physiological conditions and the observed low cytotoxicity 48 h post-transfection demonstrated the high potential of this new system for gene silencing therapy application in clinics.
Collapse
Affiliation(s)
- Diana Diaz-Dussan
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton T6G 1H9, Alberta Canada
| | - Yasuhiro Nakagawa
- International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS), Ibaraki, Japan and Graduate School of Pure and Applied Science, University of Tsukuba, Ibaraki, Japan
| | - Yi-Yang Peng
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton T6G 1H9, Alberta Canada
| | - Leslie V. Sanchez C
- Department of Chemical and Environmental Engineering, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Mitsuhiro Ebara
- International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS), Ibaraki, Japan and Graduate School of Pure and Applied Science, University of Tsukuba, Ibaraki, Japan
| | - Piyush Kumar
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, T6G 1Z2, Alberta, Canada
| | - Ravin Narain
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton T6G 1H9, Alberta Canada
| |
Collapse
|
604
|
Chow MYT, Qiu Y, Lo FFK, Lin HHS, Chan HK, Kwok PCL, Lam JKW. Inhaled powder formulation of naked siRNA using spray drying technology with l-leucine as dispersion enhancer. Int J Pharm 2017; 530:40-52. [PMID: 28720537 DOI: 10.1016/j.ijpharm.2017.07.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 06/14/2017] [Accepted: 07/05/2017] [Indexed: 01/25/2023]
Abstract
Pulmonary delivery of short interfering RNA (siRNA) has been widely studied in both animal and clinical studies to treat various respiratory diseases by gene silencing through RNA interference. Some of these studies showed that the administration of naked siRNA (without the use of any delivery vectors) could achieve satisfactory gene silencing effect, a unique feature to pulmonary delivery. Liquid aerosols were mostly used with very limited studies on the use of powder aerosols for siRNA. In this study, siRNA was co-spray dried with mannitol and l-leucine, the latter being a dispersion enhancer. To the best of our knowledge, this is the first time that siRNA in its naked form was formulated into an inhalable dry powder using spray drying technology. The aerosol performance of the powder was evaluated by Next Generation Impactor (NGI). The presence of l-leucine in the formulation could improve the aerosolization of siRNA-containing powders. Results from the X-ray photoelectron spectroscopy (XPS) suggested that l-leucine was enriched on the particle surface and promote powder dispersion. Among the different siRNA formulations being examined, the one that contained 50% w/w of l-leucine exhibited the best aerodynamic performance, with a high emitted fraction (EF) of around 80% and a modest fine particle fraction (FPF) of 45%. Importantly, the integrity of siRNA was successfully retained as evaluated by gel retardation assay and high performance liquid chromatography (HPLC).
Collapse
Affiliation(s)
- Michael Y T Chow
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Yingshan Qiu
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Fiona F K Lo
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Hinson H S Lin
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Hak-Kim Chan
- Advanced Drug Delivery Group, Faculty of Pharmacy, Building A15, The University of Sydney, Sydney, NSW 2006, Australia
| | - Philip C L Kwok
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong; Advanced Drug Delivery Group, Faculty of Pharmacy, Building A15, The University of Sydney, Sydney, NSW 2006, Australia
| | - Jenny K W Lam
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong.
| |
Collapse
|
605
|
Controlled and sustained delivery of siRNA/NPs from hydrogels expedites bone fracture healing. Biomaterials 2017; 139:127-138. [PMID: 28601703 DOI: 10.1016/j.biomaterials.2017.06.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 05/16/2017] [Accepted: 06/02/2017] [Indexed: 01/01/2023]
Abstract
Despite great potential, delivery remains as the most significant barrier to the widespread use of siRNA therapeutics. siRNA has delivery limitations due to susceptibility to RNase degradation, low cellular uptake, and poor tissue-specific localization. Here, we report the development of a hybrid nanoparticle (NP)/hydrogel system that overcomes these challenges. Hydrogels provide localized and sustained delivery via controlled release of entrapped siRNA/NP complexes while NPs protect and enable efficient cytosolic accumulation of siRNA. To demonstrate therapeutic efficacy, regenerative siRNA against WW domain-containing E3 ubiquitin protein ligase 1 (Wwp1) complexed with NP were entrapped within poly(ethylene glycol) (PEG)-based hydrogels and implanted at sites of murine mid-diaphyseal femur fractures. Results showed localization of hydrogels and controlled release of siRNA/NPs at fractures for 28 days, a timeframe over which fracture healing occurs. siRNA/NP sustained delivery from hydrogels resulted in significant Wwp1 silencing at fracture callus compared to untreated controls. Fractures treated with siRNA/NP hydrogels exhibited accelerated bone formation and significantly increased biomechanical strength. This NP/hydrogel siRNA delivery system has outstanding therapeutic promise to augment fracture healing. Owing to the structural similarities of siRNA, the development of the hydrogel platform for in vivo siRNA delivery has myriad therapeutic possibilities in orthopaedics and beyond.
Collapse
|
606
|
Wang X, Yan N, Song T, Wang B, Wei B, Lin L, Chen X, Tian H, Liang H. Robust Fuel Catalyzed DNA Molecular Machine for in Vivo MicroRNA Detection. ACTA ACUST UNITED AC 2017. [DOI: 10.1002/adbi.201700060] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Xiaojing Wang
- CAS Key Laboratory of Soft Matter Chemistry; Collaborative Innovation Center of Chemistry for Energy Materials (iChEM); Department of Polymer Science and Engineering; University of Science and Technology of China; Hefei Anhui 230026 P. R. China
| | - Nan Yan
- Key Laboratory of Polymer Ecomaterials; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 P. R. China
| | - Tingjie Song
- CAS Key Laboratory of Soft Matter Chemistry; Collaborative Innovation Center of Chemistry for Energy Materials (iChEM); Department of Polymer Science and Engineering; University of Science and Technology of China; Hefei Anhui 230026 P. R. China
| | - Bei Wang
- CAS Key Laboratory of Soft Matter Chemistry; Collaborative Innovation Center of Chemistry for Energy Materials (iChEM); Department of Polymer Science and Engineering; University of Science and Technology of China; Hefei Anhui 230026 P. R. China
| | - Bing Wei
- Hefei National Laboratory for Physical Sciences at Microscale; University of Science and Technology of China; Hefei Anhui 230026 P. R. China
| | - Lin Lin
- Key Laboratory of Polymer Ecomaterials; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 P. R. China
| | - Huayu Tian
- Key Laboratory of Polymer Ecomaterials; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 P. R. China
| | - Haojun Liang
- CAS Key Laboratory of Soft Matter Chemistry; Collaborative Innovation Center of Chemistry for Energy Materials (iChEM); Department of Polymer Science and Engineering; University of Science and Technology of China; Hefei Anhui 230026 P. R. China
- Hefei National Laboratory for Physical Sciences at Microscale; University of Science and Technology of China; Hefei Anhui 230026 P. R. China
| |
Collapse
|
607
|
Colorectal Cancer: From the Genetic Model to Posttranscriptional Regulation by Noncoding RNAs. BIOMED RESEARCH INTERNATIONAL 2017; 2017:7354260. [PMID: 28573140 PMCID: PMC5442347 DOI: 10.1155/2017/7354260] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 02/16/2017] [Indexed: 12/11/2022]
Abstract
Colorectal cancer is the third most common form of cancer in developed countries and, despite the improvements achieved in its treatment options, remains as one of the main causes of cancer-related death. In this review, we first focus on colorectal carcinogenesis and on the genetic and epigenetic alterations involved. In addition, noncoding RNAs have been shown to be important regulators of gene expression. We present a general overview of what is known about these molecules and their role and dysregulation in cancer, with a special focus on the biogenesis, characteristics, and function of microRNAs. These molecules are important regulators of carcinogenesis, progression, invasion, angiogenesis, and metastases in cancer, including colorectal cancer. For this reason, miRNAs can be used as potential biomarkers for diagnosis, prognosis, and efficacy of chemotherapeutic treatments, or even as therapeutic agents, or as targets by themselves. Thus, this review highlights the importance of miRNAs in the development, progression, diagnosis, and therapy of colorectal cancer and summarizes current therapeutic approaches for the treatment of colorectal cancer.
Collapse
|
608
|
Selective targeting of point-mutated KRAS through artificial microRNAs. Proc Natl Acad Sci U S A 2017; 114:E4203-E4212. [PMID: 28484014 DOI: 10.1073/pnas.1620562114] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Mutated protein-coding genes drive the molecular pathogenesis of many diseases, including cancer. Specifically, mutated KRAS is a documented driver for malignant transformation, occurring early during the pathogenesis of cancers such as lung and pancreatic adenocarcinomas. Therapeutically, the indiscriminate targeting of wild-type and point-mutated transcripts represents an important limitation. Here, we leveraged on the design of miRNA-like artificial molecules (amiRNAs) to specifically target point-mutated genes, such as KRAS, without affecting their wild-type counterparts. Compared with an siRNA-like approach, the requirement of perfect complementarity of the microRNA seed region to a given target sequence in the microRNA/target model has proven to be a more efficient strategy, accomplishing the selective targeting of point-mutated KRAS in vitro and in vivo.
Collapse
|
609
|
Affiliation(s)
- Masato Ikeda
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, Gifu 501-1193
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu 501-1193
- Center for Highly Advanced Integration of Nano and Life Sciences, Gifu University (G-CHAIN), Gifu 501-1193
| | - Marina Kabumoto
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu 501-1193
| |
Collapse
|
610
|
Fukagawa S, Miyata K, Yotsumoto F, Kiyoshima C, Nam SO, Anan H, Katsuda T, Miyahara D, Murata M, Yagi H, Shirota K, Yasunaga S, Kato K, Miyamoto S. MicroRNA-135a-3p as a promising biomarker and nucleic acid therapeutic agent for ovarian cancer. Cancer Sci 2017; 108:886-896. [PMID: 28231414 PMCID: PMC5448652 DOI: 10.1111/cas.13210] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 02/11/2017] [Accepted: 02/15/2017] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer is the most lethal gynecologic malignancy. Recently, several molecularly targeted anticancer agents have been developed for ovarian cancer; however, its prognosis remains extremely poor. The development of molecularly targeted therapy, as well as companion diagnostics, is required to improve outcomes for patients with ovarian cancer. In this study, to identify microRNAs (miRNAs) involved in the progression of ovarian cancer we analyzed serum miRNAs in patients with ovarian cancer using miRNA array and quantitative RT-PCR and examined the anticancer properties of miRNA expression in ovarian cancer cells. In patients with ovarian cancer, high amount of miR-135a-3p in serum samples was significantly associated with favorable clinical prognosis. The amount of miR-135a-3p was significantly decreased in patients with ovarian cancer compared with patients with ovarian cysts or normal ovaries. In SKOV-3 and ES-2 human ovarian cancer cells, enhanced expression of miR-135a-3p induced drug sensitivity to cisplatin and paclitaxel and suppressed cell proliferation and xenograft tumor growth. These findings suggest that miR-135a-3p may be considered as a biomarker and a therapeutic agent in ovarian cancer.
Collapse
Affiliation(s)
- Satoshi Fukagawa
- Department of Obstetrics and GynecologyFukuoka UniversityFukuokaJapan
- Central Research Institute for Advanced Molecular MedicineFukuoka UniversityFukuokaJapan
| | - Kohei Miyata
- Department of Obstetrics and GynecologyFukuoka UniversityFukuokaJapan
- Central Research Institute for Advanced Molecular MedicineFukuoka UniversityFukuokaJapan
| | | | - Chihiro Kiyoshima
- Department of Obstetrics and GynecologyFukuoka UniversityFukuokaJapan
- Central Research Institute for Advanced Molecular MedicineFukuoka UniversityFukuokaJapan
| | - Sung Ouk Nam
- Department of Obstetrics and GynecologyFukuoka UniversityFukuokaJapan
| | - Haruchika Anan
- Department of Obstetrics and GynecologyFukuoka UniversityFukuokaJapan
| | - Takahiro Katsuda
- Department of Obstetrics and GynecologyFukuoka UniversityFukuokaJapan
| | - Daisuke Miyahara
- Department of Obstetrics and GynecologyFukuoka UniversityFukuokaJapan
| | - Masaharu Murata
- Department of Obstetrics and GynecologyFukuoka UniversityFukuokaJapan
| | - Hiroshi Yagi
- Department of Obstetrics and GynecologyFaculty of MedicineKyushu UniversityFukuokaJapan
| | - Kyoko Shirota
- Department of Obstetrics and GynecologyFukuoka UniversityFukuokaJapan
| | - Shin'ichiro Yasunaga
- Central Research Institute for Advanced Molecular MedicineFukuoka UniversityFukuokaJapan
- Department of BiochemistryFaculty of MedicineFukuoka UniversityFukuokaJapan
| | - Kiyoko Kato
- Department of Obstetrics and GynecologyFaculty of MedicineKyushu UniversityFukuokaJapan
| | - Shingo Miyamoto
- Department of Obstetrics and GynecologyFukuoka UniversityFukuokaJapan
| |
Collapse
|
611
|
Fernandez-Piñeiro I, Badiola I, Sanchez A. Nanocarriers for microRNA delivery in cancer medicine. Biotechnol Adv 2017; 35:350-360. [PMID: 28286148 DOI: 10.1016/j.biotechadv.2017.03.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 02/26/2017] [Accepted: 03/03/2017] [Indexed: 01/09/2023]
Affiliation(s)
- I Fernandez-Piñeiro
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Santiago de Compostela (USC), Campus Vida, 15782 Santiago de Compostela, Spain
| | - I Badiola
- Department of Cell Biology and Histology, Faculty of Medicine and Odontology, University of Basque Country, B° Sarriena, s/n, 48940 Leioa, Spain
| | - A Sanchez
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Santiago de Compostela (USC), Campus Vida, 15782 Santiago de Compostela, Spain; Genetics and Biology of the Development of Kidney Diseases Unit, Sanitary Research Institute (IDIS) of the University Hospital Complex of Santiago de Compostela (CHUS), Travesía da Choupana, s/n, 15706 Santiago de Compostela, Spain.
| |
Collapse
|
612
|
Liu Q, Zhang X, Hu X, Yuan L, Cheng J, Jiang Y, Ao Y. Emerging Roles of circRNA Related to the Mechanical Stress in Human Cartilage Degradation of Osteoarthritis. MOLECULAR THERAPY. NUCLEIC ACIDS 2017. [PMID: 28624198 PMCID: PMC5415239 DOI: 10.1016/j.omtn.2017.04.004] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Circular RNAs (circRNAs) are involved in the development of various diseases; however, knowledge on circRNAs in osteoarthritis (OA) is limited. This study aims to identify circRNA expression in different regions affected by OA and to explore the function of mechanical stress-related circRNAs (circRNAs-MSR) in cartilage. Bioinformatics was employed to predict the interaction of circRNAs and mRNAs in the cartilage. Loss-of-function experiments for circRNAs-MSR were performed in vitro. A total of 104 circRNAs were differentially expressed in damaged versus intact cartilage. Of these circRNAs, 44 and 60 were upregulated and downregulated, respectively, in the damaged tissue. circRNA-MSR expression increased under mechanical stress in chondrocytes. circRNAs-MSR were silenced using small interfering RNA, and knockdown of circRNAs-MSR could suppress tumor necrosis factor alpha (TNF-α) expression and increase extracellular matrix (ECM) formation. Our results demonstrated that circRNAs-MSR regulated TNF-α expression and participated in the chondrocyte ECM degradation process. We propose that the inhibition of circRNAs-MSR could inhibit the degradation of chondrocyte ECM and knockdown of circRNAs-MSR could be a potential therapeutic target for OA.
Collapse
Affiliation(s)
- Qiang Liu
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, People's Republic of China
| | - Xin Zhang
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, People's Republic of China
| | - Xiaoqing Hu
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, People's Republic of China
| | - Lan Yuan
- Medical and Healthy Analysis Centre, Peking University, 38 Xueyuan Road, Haidian District, Beijing 100191, People's Republic of China
| | - Jin Cheng
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, People's Republic of China
| | - Yanfang Jiang
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, People's Republic of China
| | - Yingfang Ao
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, People's Republic of China.
| |
Collapse
|
613
|
Stenfeldt C, Arzt J, Smoliga G, LaRocco M, Gutkoska J, Lawrence P. Proof-of-concept study: profile of circulating microRNAs in Bovine serum harvested during acute and persistent FMDV infection. Virol J 2017; 14:71. [PMID: 28388926 PMCID: PMC5384155 DOI: 10.1186/s12985-017-0743-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 03/29/2017] [Indexed: 02/07/2023] Open
Abstract
Background Changes in the levels of circulating microRNAs (miRNAs) in the serum of humans and animals have been detected as a result of infection with a variety of viruses. However, to date, such a miRNA profiling study has not been conducted for foot-and-mouth disease virus (FMDV) infection. Methods The relative abundance of 169 miRNAs was measured in bovine serum collected at three different phases of FMDV infection in a proof-of-concept study using miRNA PCR array plates. Results Alterations in specific miRNA levels were detected in serum during acute, persistent, and convalescent phases of FMDV infection. Subclinical FMDV persistence produced a circulating miRNA profile distinct from cattle that had cleared infection. bta-miR-17-5p was highest expressed during acute infection, whereas bta-miR-31 was the highest during FMDV persistence. Interestingly, miR-1281was significantly down-regulated during both acute and persistent infection. Cattle that cleared infection resembled the baseline profile, adding support to applying serum miRNA profiling for identification of sub-clinically infected FMDV carriers. Significantly regulated miRNAs during acute or persistent infection were associated with cellular proliferation, apoptosis, modulation of the immune response, and lipid metabolism. Conclusions These findings suggest a role for non-coding regulatory RNAs in FMDV infection of cattle. Future studies will delineate the individual contributions of the reported miRNAs to FMDV replication, determine if this miRNA signature is applicable across all FMDV serotypes, and may facilitate development of novel diagnostic applications. Electronic supplementary material The online version of this article (doi:10.1186/s12985-017-0743-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Carolina Stenfeldt
- Plum Island Animal Disease Center, USDA/ARS/NAA/FADRU, P.O. Box 848, Greenport, NY, 11944-0848, USA
| | - Jonathan Arzt
- Plum Island Animal Disease Center, USDA/ARS/NAA/FADRU, P.O. Box 848, Greenport, NY, 11944-0848, USA
| | - George Smoliga
- Plum Island Animal Disease Center, USDA/ARS/NAA/FADRU, P.O. Box 848, Greenport, NY, 11944-0848, USA
| | - Michael LaRocco
- Plum Island Animal Disease Center, USDA/ARS/NAA/FADRU, P.O. Box 848, Greenport, NY, 11944-0848, USA
| | - Joseph Gutkoska
- Plum Island Animal Disease Center, USDA/ARS/NAA/FADRU, P.O. Box 848, Greenport, NY, 11944-0848, USA
| | - Paul Lawrence
- Plum Island Animal Disease Center, USDA/ARS/NAA/FADRU, P.O. Box 848, Greenport, NY, 11944-0848, USA.
| |
Collapse
|
614
|
Bartneck M. Immunomodulatory Nanomedicine. Macromol Biosci 2017; 17. [DOI: 10.1002/mabi.201700021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 02/21/2017] [Indexed: 12/28/2022]
Affiliation(s)
- Matthias Bartneck
- Department of Medicine III; Medical Faculty; RWTH Aachen; Pauwelsstr. 30 52074 Aachen Germany
| |
Collapse
|
615
|
Indersie E, Lesjean S, Hooks KB, Sagliocco F, Ernault T, Cairo S, Merched-Sauvage M, Rullier A, Le Bail B, Taque S, Grotzer M, Branchereau S, Guettier C, Fabre M, Brugières L, Hagedorn M, Buendia MA, Grosset CF. MicroRNA therapy inhibits hepatoblastoma growth in vivo by targeting β-catenin and Wnt signaling. Hepatol Commun 2017; 1:168-183. [PMID: 29404451 PMCID: PMC5721429 DOI: 10.1002/hep4.1029] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/24/2017] [Accepted: 02/27/2017] [Indexed: 01/03/2023] Open
Abstract
Hepatoblastoma (HBL) is the most common pediatric liver cancer. In this malignant neoplasm, beta-catenin protein accumulates and increases Wnt signaling due to recurrent activating mutations in the catenin-beta 1 (CTNNB1) gene. Therefore, beta-catenin is a key therapeutic target in HBL. However, controlling beta-catenin production with therapeutic molecules has been challenging. New biological studies could provide alternative therapeutic solutions for the treatment of HBL, especially for advanced tumors and metastatic disease. In this study, we identified microRNAs (miRNAs) that target beta-catenin and block HBL cell proliferation in vitro and tumor growth in vivo. Using our dual-fluorescence-FunREG system, we screened a library of 1,712 miRNA mimics and selected candidates inhibiting CTNNB1 expression through interaction with its untranslated regions. After validating the regulatory effect of nine miRNAs on beta-catenin in HBL cells, we measured their expression in patient samples. Let-7i-3p, miR-449b-3p, miR-624-5p, and miR-885-5p were decreased in tumors compared to normal livers. Moreover, they inhibited HBL cell growth and Wnt signaling activity in vitro partly through beta-catenin down-regulation. Additionally, miR-624-5p induced cell senescence in vitro, blocked experimental HBL growth in vivo, and directly targeted the beta-catenin 3'-untranslated region. Conclusion: Our results shed light on how beta-catenin-regulating miRNAs control HBL progression through Wnt signaling inactivation. In particular, miR-624-5p may constitute a promising candidate for miRNA replacement therapy for HBL patients. (Hepatology Communications 2017;1:168-183).
Collapse
Affiliation(s)
- Emilie Indersie
- Univ. Bordeaux, Inserm, GREF, U1053, 33076 Bordeaux France.,Univ. Bordeaux, Inserm, BMGIC, U1035, 33076 Bordeaux France
| | - Sarah Lesjean
- Univ. Bordeaux, Inserm, GREF, U1053, 33076 Bordeaux France.,Univ. Bordeaux, Inserm, BMGIC, U1035, 33076 Bordeaux France
| | - Katarzyna B Hooks
- Univ. Bordeaux, Inserm, GREF, U1053, 33076 Bordeaux France.,Univ. Bordeaux, Inserm, BMGIC, U1035, 33076 Bordeaux France
| | - Francis Sagliocco
- Univ. Bordeaux, Inserm, GREF, U1053, 33076 Bordeaux France.,Univ. Bordeaux, Inserm, BMGIC, U1035, 33076 Bordeaux France
| | - Tony Ernault
- INSERM, UMR 1193, Paul-Brousse Hospital, Hepatobiliary Centre F-94800 Villejuif France.,Univ. Paris Saclay F-94800 Villejuif France
| | - Stefano Cairo
- XenTechEvry France.,Laboratory for Technologies of Advanced Therapies, Department of Morphology, Surgery and Experimental Medicine University of Ferrara Italy
| | | | | | | | | | - Michael Grotzer
- SIOPEL (International Childhood Liver Tumours Strategy Group) Liver Tumor and Tissue Banking Program University Children's Hospital Zürich Switzerland
| | | | | | | | | | - Martin Hagedorn
- Univ. Bordeaux, Inserm, GREF, U1053, 33076 Bordeaux France.,Univ. Bordeaux, Inserm, BMGIC, U1035, 33076 Bordeaux France
| | - Marie-Annick Buendia
- INSERM, UMR 1193, Paul-Brousse Hospital, Hepatobiliary Centre F-94800 Villejuif France.,Univ. Paris Saclay F-94800 Villejuif France
| | - Christophe F Grosset
- Univ. Bordeaux, Inserm, GREF, U1053, 33076 Bordeaux France.,Univ. Bordeaux, Inserm, BMGIC, U1035, 33076 Bordeaux France
| |
Collapse
|
616
|
Prabhakar B, Zhong XB, Rasmussen TP. Exploiting Long Noncoding RNAs as Pharmacological Targets to Modulate Epigenetic Diseases. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2017; 90:73-86. [PMID: 28356895 PMCID: PMC5369047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Long non-coding RNAs (lncRNAs) constitute the largest class of non-coding transcripts in the human genome. Results from next-generation sequencing and bioinformatics advances indicate that the human genome contains more non-coding RNA genes than protein-coding genes. Validated functions of lncRNAs suggest that they are master regulators of gene expression and often exert their influences via epigenetic mechanisms by modulating chromatin structure. Specific lncRNAs can regulate transcription in gene clusters. Since the functions of protein-coding genes in clusters are often tied to specific pathways, lncRNAs constitute attractive pharmacological targets. Here we review the current knowledge of lncRNA functions in human cells and their roles in disease processes. We also present forward-looking perspectives on how they might be manipulated pharmacologically for the treatment of a variety of human diseases, in which regulation of gene expression by epigenetic mechanisms plays a major role.
Collapse
Affiliation(s)
- Bindu Prabhakar
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT
| | - Xiao-bo Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT,Institute for Systems Genomics, University of Connecticut, Storrs/Farmington, CT
| | - Theodore P. Rasmussen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT,Institute for Systems Genomics, University of Connecticut, Storrs/Farmington, CT,To whom all correspondence should be addressed: Theodore P. Rasmussen, Ph.D., Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road, Unit 3092, Storrs, CT 06269; Tel: (860) 486-8339; Fax: (860) 486-5792;
| |
Collapse
|
617
|
Ouyang M, Hill W, Lee JH, Hur SC. Microscale Symmetrical Electroporator Array as a Versatile Molecular Delivery System. Sci Rep 2017; 7:44757. [PMID: 28317836 PMCID: PMC5357946 DOI: 10.1038/srep44757] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 02/13/2017] [Indexed: 12/13/2022] Open
Abstract
Successful developments of new therapeutic strategies often rely on the ability to deliver exogenous molecules into cytosol. We have developed a versatile on-chip vortex-assisted electroporation system, engineered to conduct sequential intracellular delivery of multiple molecules into various cell types at low voltage in a dosage-controlled manner. Micro-patterned planar electrodes permit substantial reduction in operational voltages and seamless integration with an existing microfluidic technology. Equipped with real-time process visualization functionality, the system enables on-chip optimization of electroporation parameters for cells with varying properties. Moreover, the system’s dosage control and multi-molecular delivery capabilities facilitate intracellular delivery of various molecules as a single agent or in combination and its utility in biological research has been demonstrated by conducting RNA interference assays. We envision the system to be a powerful tool, aiding a wide range of applications, requiring single-cell level co-administrations of multiple molecules with controlled dosages.
Collapse
Affiliation(s)
- Mengxing Ouyang
- Rowland Institute at Harvard University, 100 Edwin H. Land Blvd., Cambridge, MA 02142, USA
| | - Winfield Hill
- Rowland Institute at Harvard University, 100 Edwin H. Land Blvd., Cambridge, MA 02142, USA
| | - Jung Hyun Lee
- Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Soojung Claire Hur
- Rowland Institute at Harvard University, 100 Edwin H. Land Blvd., Cambridge, MA 02142, USA
| |
Collapse
|
618
|
Local co-administration of gene-silencing RNA and drugs in cancer therapy: State-of-the art and therapeutic potential. Cancer Treat Rev 2017; 55:128-135. [PMID: 28363142 DOI: 10.1016/j.ctrv.2017.03.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 03/06/2017] [Accepted: 03/07/2017] [Indexed: 12/12/2022]
Abstract
Gene-silencing miRNA and siRNA are emerging as attractive therapeutics with potential to suppress any genes, which could be especially useful in combination cancer therapy to overcome multidrug resistant (MDR) cancer. Nanomedicine aims to advance cancer treatment through functional nanocarriers that delivers one or more therapeutics to cancer tissue and cells with minimal off-target effects and suitable release kinetics and dosages. Although much effort has gone into developing circulating nanocarriers with targeting functionality for systemic administration, another alternative and straightforward approach is to utilize formulations to be administered directly to the site of action, such as pulmonary and intratumoral delivery. The combination of gene-silencing RNA with drugs in nanocarriers for localized delivery is emerging with promising results. In this review, the current progress and strategies for local co-administration of RNA and drug for synergistic effects and future potential in cancer treatment are presented and discussed. Key advances in RNA-drug anticancer synergy and localized delivery systems were combined with a review of the available literature on local co-administration of RNA and drug for cancer treatment. It is concluded that advanced delivery systems for local administration of gene-silencing RNA and drug hold potential in treatment of cancer, depending on indication. In particular, there are promising developments using pulmonary delivery and intratumoral delivery in murine models, but further research should be conducted on other local administration strategies, designs that achieve effective intracellular delivery and maximize synergy and feasibility for clinical use.
Collapse
|
619
|
Mendes R, Fernandes AR, Baptista PV. Gold Nanoparticle Approach to the Selective Delivery of Gene Silencing in Cancer-The Case for Combined Delivery? Genes (Basel) 2017; 8:E94. [PMID: 28257109 PMCID: PMC5368698 DOI: 10.3390/genes8030094] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/25/2017] [Accepted: 02/23/2017] [Indexed: 01/30/2023] Open
Abstract
Gene therapy arises as a great promise for cancer therapeutics due to its potential to silence genes involved in tumor development. In fact, there are some pivotal gene drivers that suffer critical alterations leading to cell transformation and ultimately to tumor growth. In this vein, gene silencing has been proposed as an active tool to selectively silence these molecular triggers of cancer, thus improving treatment. However, naked nucleic acid (DNA/RNA) sequences are reported to have a short lifetime in the body, promptly degraded by circulating enzymes, which in turn speed up elimination and decrease the therapeutic potential of these drugs. The use of nanoparticles for the effective delivery of these silencers to the specific target locations has allowed researchers to overcome this issue. Particularly, gold nanoparticles (AuNPs) have been used as attractive vehicles for the target-specific delivery of gene-silencing moieties, alone or in combination with other drugs. We shall discuss current trends in AuNP-based delivery of gene-silencing tools, considering the promising road ahead without overlooking existing concerns for their translation to clinics.
Collapse
Affiliation(s)
- Rita Mendes
- UCIBIO, DCV, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal.
| | - Alexandra R Fernandes
- UCIBIO, DCV, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal.
| | - Pedro V Baptista
- UCIBIO, DCV, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal.
| |
Collapse
|
620
|
Mihailovic MK, Chen A, Gonzalez-Rivera JC, Contreras LM. Defective Ribonucleoproteins, Mistakes in RNA Processing, and Diseases. Biochemistry 2017; 56:1367-1382. [PMID: 28206738 DOI: 10.1021/acs.biochem.6b01134] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Ribonucleoproteins (RNPs) are vital to many cellular events. To this end, many neurodegenerative diseases and cancers have been linked to RNP malfunction, particularly as this relates to defective processing of cellular RNA. The connection of RNPs and diseases has also propagated a shift of focus onto RNA targeting from traditional protein targeting treatments. However, therapeutic development in this area has been limited by incomplete molecular insight into the specific contributions of RNPs to disease. This review outlines the role of several RNPs in diseases, focusing on molecular defects in processes that affect proper RNA handling in the cell. This work also evaluates the contributions of recently developed methods to understanding RNP association and function. We review progress in this area by focusing on molecular malfunctions of RNPs associated with the onset and progression of several neurodegenerative diseases and cancer and conclude with a brief discussion of RNA-based therapeutic efforts.
Collapse
Affiliation(s)
- Mia K Mihailovic
- McKetta Department of Chemical Engineering, University of Texas at Austin , 200 East. Dean Keeton Street, Stop C0400, Austin, Texas 78712, United States
| | - Angela Chen
- McKetta Department of Chemical Engineering, University of Texas at Austin , 200 East. Dean Keeton Street, Stop C0400, Austin, Texas 78712, United States
| | - Juan C Gonzalez-Rivera
- McKetta Department of Chemical Engineering, University of Texas at Austin , 200 East. Dean Keeton Street, Stop C0400, Austin, Texas 78712, United States
| | - Lydia M Contreras
- McKetta Department of Chemical Engineering, University of Texas at Austin , 200 East. Dean Keeton Street, Stop C0400, Austin, Texas 78712, United States
| |
Collapse
|
621
|
Obora K, Onodera Y, Takehara T, Frampton J, Hasei J, Ozaki T, Teramura T, Fukuda K. Inflammation-induced miRNA-155 inhibits self-renewal of neural stem cells via suppression of CCAAT/enhancer binding protein β (C/EBPβ) expression. Sci Rep 2017; 7:43604. [PMID: 28240738 PMCID: PMC5378916 DOI: 10.1038/srep43604] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/24/2017] [Indexed: 01/04/2023] Open
Abstract
Intracerebral inflammation resulting from injury or disease is implicated in disruption of neural regeneration and may lead to irreversible neuronal dysfunction. Analysis of inflammation-related microRNA profiles in various tissues, including the brain, has identified miR-155 among the most prominent miRNAs linked to inflammation. Here, we hypothesize that miR-155 mediates inflammation-induced suppression of neural stem cell (NSC) self-renewal. Using primary mouse NSCs and human NSCs derived from induced pluripotent stem (iPS) cells, we demonstrate that three important genes involved in NSC self-renewal (Msi1, Hes1 and Bmi1) are suppressed by miR-155. We also demonstrate that suppression of self-renewal genes is mediated by the common transcription factor C/EBPβ, which is a direct target of miR-155. Our study describes an axis linking inflammation and miR-155 to expression of genes related to NSC self-renewal, suggesting that regulation of miR-155 may hold potential as a novel therapeutic strategy for treating neuroinflammatory diseases.
Collapse
Affiliation(s)
- Kayoko Obora
- Department of Rehabilitation Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| | - Yuta Onodera
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| | - Toshiyuki Takehara
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| | - John Frampton
- School of Biomedical Engineering, Dalhousie University. Halifax, Nova Scotia, Canada
| | - Joe Hasei
- Science of Functional Recovery and Reconstruction, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toshifumi Ozaki
- Science of Functional Recovery and Reconstruction, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takeshi Teramura
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| | - Kanji Fukuda
- Department of Rehabilitation Medicine, Kindai University Faculty of Medicine, Osaka, Japan.,Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| |
Collapse
|
622
|
Antimisiaris S, Mourtas S, Papadia K. Targeted si-RNA with liposomes and exosomes (extracellular vesicles): How to unlock the potential. Int J Pharm 2017; 525:293-312. [PMID: 28163221 DOI: 10.1016/j.ijpharm.2017.01.056] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 01/25/2017] [Accepted: 01/27/2017] [Indexed: 12/17/2022]
Abstract
The concept of RNA interference therapeutics has been initiated 18 years ago, and the main bottleneck for translation of the technology into therapeutic products remains the delivery of functional RNA molecules into the cell cytoplasm. In the present review article after an introduction about the theoretical basis of RNAi therapy and the main challenges encountered for its realization, an overview of the different types of delivery systems or carriers, used as potential systems to overcome RNAi delivery issues, will be provided. Characteristic examples or results obtained with the most promising systems will be discussed. Focus will be given mostly on the applications of liposomes or other types of lipid carriers, such as exosomes, towards improved delivery of RNAi to therapeutic targets. Finally the approach of integrating the advantages of these two vesicular systems, liposomes and exosomes, as a potential solution to realize RNAi therapy, will be proposed.
Collapse
Affiliation(s)
- Sophia Antimisiaris
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, University of Patras, Rio 26504, Greece; Institute of Chemical Engineering, FORTH/ICE-HT, Rio 26504, Greece.
| | - Spyridon Mourtas
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, University of Patras, Rio 26504, Greece
| | - Konstantina Papadia
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, University of Patras, Rio 26504, Greece
| |
Collapse
|
623
|
Fukuda M, Umeno H, Nose K, Nishitarumizu A, Noguchi R, Nakagawa H. Construction of a guide-RNA for site-directed RNA mutagenesis utilising intracellular A-to-I RNA editing. Sci Rep 2017; 7:41478. [PMID: 28148949 PMCID: PMC5288656 DOI: 10.1038/srep41478] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 12/20/2016] [Indexed: 01/24/2023] Open
Abstract
As an alternative to DNA mutagenesis, RNA mutagenesis can potentially become a powerful gene-regulation method for fundamental research and applied life sciences. Adenosine-to-inosine (A-to-I) RNA editing alters genetic information at the transcript level and is an important biological process that is commonly conserved in metazoans. Therefore, a versatile RNA-mutagenesis method can be achieved by utilising the intracellular RNA-editing mechanism. Here, we report novel guide RNAs capable of inducing A-to-I mutations by guiding the editing enzyme, human adenosine deaminase acting on RNA (ADAR). These guide RNAs successfully introduced A-to-I mutations into the target-site, which was determined by the reprogrammable antisense region. In ADAR2-over expressing cells, site-directed RNA editing could also be performed by simply introducing the guide RNA. Our guide RNA framework provides basic insights into establishing a generally applicable RNA-mutagenesis method.
Collapse
Affiliation(s)
- Masatora Fukuda
- Department of Chemistry, Faculty of Science, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, Fukuoka 814-0180, Japan
| | - Hiromitsu Umeno
- Department of Chemistry, Faculty of Science, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, Fukuoka 814-0180, Japan
| | - Kanako Nose
- Department of Chemistry, Faculty of Science, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, Fukuoka 814-0180, Japan
| | - Azusa Nishitarumizu
- Department of Chemistry, Faculty of Science, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, Fukuoka 814-0180, Japan
| | - Ryoma Noguchi
- Department of Chemistry, Faculty of Science, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, Fukuoka 814-0180, Japan
| | - Hiroyuki Nakagawa
- Department of Earth System Science, Faculty of Science, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, Fukuoka, 814-0180, Japan
| |
Collapse
|
624
|
Gutkoska J, LaRocco M, Ramirez-Medina E, de Los Santos T, Lawrence P. Host microRNA-203a Is antagonistic to the progression of foot-and-mouth disease virus infection. Virology 2017; 504:52-62. [PMID: 28152384 DOI: 10.1016/j.virol.2017.01.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/22/2017] [Accepted: 01/23/2017] [Indexed: 12/11/2022]
Abstract
Sam68 was previously shown to be a critical host factor for foot-and-mouth disease virus (FMDV) replication. MicroRNA (miR) miR-203a is reportedly a negative regulator of Sam68 expression both in vitro and in vivo. Here, transfection of miR-203a-3p and miR-203a-5p mimics separately and in combination in a porcine cell line followed by FMDV infection resulted in diminished viral protein synthesis and a 4 and 6log reduction in virus titers relative to negative controls, respectively. Unexpectedly, Sam68 expression was increased by miR-203a-5p transfection, but not miR-203a-3p. miR-203a-5p also down-regulated Survivin expression, which was predicted to play a role in FMDV infection. Moreover, miR-203a-5p but not miR-203a-3p affected a reduction in FMDV viral RNA. These effects were not replicated with a related Picornavirus, suggesting FMDV specificity. Importantly, miR-203a-3p and miR-203a-5p impaired FMDV infection across multiple FMDV serotypes. We concluded that miR-203a-3p and miR-203a-5p represent attractive potential naturally occurring bio-therapeutics against FMDV.
Collapse
Affiliation(s)
- Joseph Gutkoska
- Plum Island Animal Disease Center Foreign Animal Disease Research Unit (FADRU) Agricultural Research Service (ARS), United States Department of Agriculture (USDA), 40550 Route 25, Orient Point, NY 11957, United States
| | - Michael LaRocco
- Plum Island Animal Disease Center Foreign Animal Disease Research Unit (FADRU) Agricultural Research Service (ARS), United States Department of Agriculture (USDA), 40550 Route 25, Orient Point, NY 11957, United States
| | - Elizabeth Ramirez-Medina
- Plum Island Animal Disease Center Foreign Animal Disease Research Unit (FADRU) Agricultural Research Service (ARS), United States Department of Agriculture (USDA), 40550 Route 25, Orient Point, NY 11957, United States
| | - Teresa de Los Santos
- Plum Island Animal Disease Center Foreign Animal Disease Research Unit (FADRU) Agricultural Research Service (ARS), United States Department of Agriculture (USDA), 40550 Route 25, Orient Point, NY 11957, United States
| | - Paul Lawrence
- Plum Island Animal Disease Center Foreign Animal Disease Research Unit (FADRU) Agricultural Research Service (ARS), United States Department of Agriculture (USDA), 40550 Route 25, Orient Point, NY 11957, United States.
| |
Collapse
|
625
|
Zhang RX, Ahmed T, Li LY, Li J, Abbasi AZ, Wu XY. Design of nanocarriers for nanoscale drug delivery to enhance cancer treatment using hybrid polymer and lipid building blocks. NANOSCALE 2017; 9:1334-1355. [PMID: 27973629 DOI: 10.1039/c6nr08486a] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Polymer-lipid hybrid nanoparticles (PLN) are an emerging nanocarrier platform made from building blocks of polymers and lipids. PLN integrate the advantages of biomimetic lipid-based nanoparticles (i.e. solid lipid nanoparticles and liposomes) and biocompatible polymeric nanoparticles. PLN are constructed from diverse polymers and lipids and their numerous combinations, which imparts PLN with great versatility for delivering drugs of various properties to their nanoscale targets. PLN can be classified into two types based on their hybrid nanoscopic structure and assembly methods: Type-I monolithic matrix and Type-II core-shell systems. This article reviews the history of PLN development, types of PLN, lipid and polymer candidates, fabrication methods, and unique properties of PLN. The applications of PLN in delivery of therapeutic or imaging agents alone or in combination for cancer treatment are summarized and illustrated with examples. Important considerations for the rational design of PLN for advanced nanoscale drug delivery are discussed, including selection of excipients, synthesis processes governing formulation parameters, optimization of nanoparticle properties, improvement of particle surface functionality to overcome macroscopic, microscopic and cellular biological barriers. Future directions and potential clinical translation of PLN are also suggested.
Collapse
Affiliation(s)
- Rui Xue Zhang
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, CanadaM5S 3M2.
| | - Taksim Ahmed
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, CanadaM5S 3M2.
| | - Lily Yi Li
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, CanadaM5S 3M2.
| | - Jason Li
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, CanadaM5S 3M2.
| | - Azhar Z Abbasi
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, CanadaM5S 3M2.
| | - Xiao Yu Wu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, CanadaM5S 3M2.
| |
Collapse
|
626
|
Nguyen MK, McMillan A, Huynh CT, Schapira DS, Alsberg E. Photocrosslinkable, biodegradable hydrogels with controlled cell adhesivity for prolonged siRNA delivery to hMSCs to enhance their osteogenic differentiation. J Mater Chem B 2017; 5:485-495. [PMID: 28652917 PMCID: PMC5482539 DOI: 10.1039/c6tb01739h] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Photocrosslinked, biodegradable hydrogels have been extensively investigated for biomedical applications, including drug delivery and tissue engineering. Here, dextran (DEX) was chemically modified with mono(2-acryloyloxyethyl) succinate (MAES) via an esterification reaction, resulting in macromers that could be photocrosslinked to form hydrolytically degradable hydrogels. Hydrogel swelling ratio and degradation rate were controlled by varying the degree of MAES modification. Thiolated cell adhesion peptides (GRGDSPC) were conjugated to acrylated dextran via thiol-acrylate reaction to regulate the interactions of human mesenchymal stem cells (hMSCs) with the photocrosslinkable hydrogels. The hydrogels permitted sustained release of short interfering RNA (siRNA) over 7 weeks and were cytocompatible with hMSCs. Sustained presentation of siRNA from these photocrosslinked DEX hydrogels enhanced the osteogenic differentiation of encapsulated hMSCs. These DEX hydrogels with tunable siRNA delivery and cell adhesive properties may provide an excellent platform for bioactive molecule delivery and tissue regeneration applications.
Collapse
Affiliation(s)
- Minh Khanh Nguyen
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106
| | - Alexandra McMillan
- Department of Pathology, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106
| | - Cong Truc Huynh
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106
| | - Daniel S Schapira
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106
- Department of Orthopaedic Surgery, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106
- National Center for Regenerative Medicine, Division of General Medical Sciences, Case Western Reserve University, 10900 Euclid Ave., Cleveland, Ohio 44106
| |
Collapse
|
627
|
Wang LL, Burdick JA. Engineered Hydrogels for Local and Sustained Delivery of RNA-Interference Therapies. Adv Healthc Mater 2017; 6:10.1002/adhm.201601041. [PMID: 27976524 PMCID: PMC5226889 DOI: 10.1002/adhm.201601041] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/21/2016] [Indexed: 12/20/2022]
Abstract
It has been nearly two decades since RNA-interference (RNAi) was first reported. While there are no approved clinical uses, several phase II and III clinical trials suggest the great promise of RNAi therapeutics. One challenge for RNAi therapies is the controlled localization and sustained presentation to target tissues, to both overcome systemic toxicity concerns and to enhance in vivo efficacy. One approach that is emerging to address these limitations is the entrapment of RNAi molecules within hydrogels for local and sustained release. In these systems, nucleic acids are either delivered as siRNA conjugates or within nanoparticles. A plethora of hydrogels has been implemented using these approaches, including both traditional hydrogels that have already been developed for other applications and new hydrogels developed specifically for RNAi delivery. These hydrogels have been applied to various applications in vivo, including cancer, bone regeneration, inflammation and cardiac repair. This review will examine the design and implementation of such hydrogel RNAi systems and will cover the most recent applications of these systems.
Collapse
Affiliation(s)
- Leo L. Wang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
628
|
Chin WX, Ang SK, Chu JJH. Recent advances in therapeutic recruitment of mammalian RNAi and bacterial CRISPR-Cas DNA interference pathways as emerging antiviral strategies. Drug Discov Today 2017; 22:17-30. [DOI: 10.1016/j.drudis.2016.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 07/20/2016] [Accepted: 08/19/2016] [Indexed: 01/01/2023]
|
629
|
Nagpal G, Chaudhary K, Dhanda SK, Raghava GPS. Computational Prediction of the Immunomodulatory Potential of RNA Sequences. Methods Mol Biol 2017; 1632:75-90. [PMID: 28730433 DOI: 10.1007/978-1-4939-7138-1_5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Advances in the knowledge of various roles played by non-coding RNAs have stimulated the application of RNA molecules as therapeutics. Among these molecules, miRNA, siRNA, and CRISPR-Cas9 associated gRNA have been identified as the most potent RNA molecule classes with diverse therapeutic applications. One of the major limitations of RNA-based therapeutics is immunotoxicity of RNA molecules as it may induce the innate immune system. In contrast, RNA molecules that are potent immunostimulators are strong candidates for use in vaccine adjuvants. Thus, it is important to understand the immunotoxic or immunostimulatory potential of these RNA molecules. The experimental techniques for determining immunostimulatory potential of siRNAs are time- and resource-consuming. To overcome this limitation, recently our group has developed a web-based server "imRNA" for predicting the immunomodulatory potential of RNA sequences. This server integrates a number of modules that allow users to perform various tasks including (1) generation of RNA analogs with reduced immunotoxicity, (2) identification of highly immunostimulatory regions in RNA sequence, and (3) virtual screening. This server may also assist users in the identification of minimum mutations required in a given RNA sequence to minimize its immunomodulatory potential that is required for designing RNA-based therapeutics. Besides, the server can be used for designing RNA-based vaccine adjuvants as it may assist users in the identification of mutations required for increasing immunomodulatory potential of a given RNA sequence. In summary, this chapter describes major applications of the "imRNA" server in designing RNA-based therapeutics and vaccine adjuvants (http://www.imtech.res.in/raghava/imrna/).
Collapse
Affiliation(s)
- Gandharva Nagpal
- Bioinformatics Centre, Institute of Microbial Technology, Sec 39A, Chandigarh, India
| | - Kumardeep Chaudhary
- Bioinformatics Centre, Institute of Microbial Technology, Sec 39A, Chandigarh, India
| | - Sandeep Kumar Dhanda
- Bioinformatics Centre, Institute of Microbial Technology, Sec 39A, Chandigarh, India
| | | |
Collapse
|
630
|
Abstract
Physiological characteristics of diseases bring about both challenges and opportunities for targeted drug delivery. Various drug delivery platforms have been devised ranging from macro- to micro- and further into the nanoscopic scale in the past decades. Recently, the favorable physicochemical properties of nanomaterials, including long circulation, robust tissue and cell penetration attract broad interest, leading to extensive studies for therapeutic benefits. Accumulated knowledge about the physiological barriers that affect the in vivo fate of nanomedicine has led to more rational guidelines for tailoring the nanocarriers, such as size, shape, charge, and surface ligands. Meanwhile, progresses in material chemistry and molecular pharmaceutics generate a panel of physiological stimuli-responsive modules that are equipped into the formulations to prepare “smart” drug delivery systems. The capability of harnessing physiological traits of diseased tissues to control the accumulation of or drug release from nanomedicine has further improved the controlled drug release profiles with a precise manner. Successful clinical translation of a few nano-formulations has excited the collaborative efforts from the research community, pharmaceutical industry, and the public towards a promising future of smart drug delivery.
Collapse
Affiliation(s)
- Wujin Sun
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Quanyin Hu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Wenyan Ji
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Grace Wright
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Zhen Gu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
631
|
Denizli M, Aslan B, Mangala LS, Jiang D, Rodriguez-Aguayo C, Lopez-Berestein G, Sood AK. Chitosan Nanoparticles for miRNA Delivery. Methods Mol Biol 2017; 1632:219-230. [PMID: 28730442 PMCID: PMC7423176 DOI: 10.1007/978-1-4939-7138-1_14] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
RNA interference techniques represent a promising strategy for therapeutic applications. In addition to small interfering RNA-based approaches, which have been widely studied and translated into clinical investigations, microRNA-based approaches are attractive owing to their "one hit, multiple targets" concept. To overcome challenges with in vivo delivery of microRNAs related to stability, cellular uptake, and specific delivery, our group has developed and characterized chitosan nanoparticles for nucleotide delivery. This platform allows for robust target modulation and antitumor activity following intravenous administration.
Collapse
Affiliation(s)
- Merve Denizli
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Burcu Aslan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for RNA Interference and Non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lingegowda S Mangala
- Center for RNA Interference and Non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Dan L. Duncan Bldg., CPB6.3275, 1515 Holcombe Blvd., Unit 1362, Houston, TX, 77030, USA
| | - Dahai Jiang
- Center for RNA Interference and Non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Dan L. Duncan Bldg., CPB6.3275, 1515 Holcombe Blvd., Unit 1362, Houston, TX, 77030, USA
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for RNA Interference and Non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for RNA Interference and Non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anil K Sood
- Center for RNA Interference and Non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Dan L. Duncan Bldg., CPB6.3275, 1515 Holcombe Blvd., Unit 1362, Houston, TX, 77030, USA.
- Program in Cancer Biology and Cancer Metastasis, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA.
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
632
|
Biodegradable nano-polymers as delivery vehicles for therapeutic small non-coding ribonucleic acids. J Control Release 2017; 245:116-126. [DOI: 10.1016/j.jconrel.2016.11.017] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 11/14/2016] [Indexed: 12/20/2022]
|
633
|
Zhang J, Cai K. Integration of polymers in the pore space of mesoporous nanocarriers for drug delivery. J Mater Chem B 2017; 5:8891-8903. [DOI: 10.1039/c7tb02559a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The construction of carrier-polymer–drug hybrids in confined nanopore space is reviewed for advancing related drug delivery systems.
Collapse
Affiliation(s)
- Jixi Zhang
- Key Laboratory of Biorheological Science and Technology
- Ministry of Education
- College of Bioengineering
- Chongqing University
- Chongqing 400044
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology
- Ministry of Education
- College of Bioengineering
- Chongqing University
- Chongqing 400044
| |
Collapse
|
634
|
Lukasik A, Zielenkiewicz P. Plant MicroRNAs-Novel Players in Natural Medicine? Int J Mol Sci 2016; 18:ijms18010009. [PMID: 28025496 PMCID: PMC5297644 DOI: 10.3390/ijms18010009] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 12/14/2016] [Accepted: 12/16/2016] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) represent a class of small non-coding RNAs that act as efficient gene expression regulators and thus play many important roles in living organisms. Due to their involvement in several known human pathological and pathogenic states, miRNA molecules have become an important issue in medicine and gained the attention of scientists from the pharmaceutical industry. In recent few years, a growing number of studies have provided evidence that miRNAs may be transferred from one species to another and regulate gene expression in the recipients’ cells. The most intriguing results revealed that stable miRNAs derived from food plants may enter the mammals’ circulatory system and, after reaching the target, inhibit the production of specific mammalian protein. Part of the scientific community has perceived this as an attractive hypothesis that may provide a foundation for novel therapeutic approaches. In turn, others are convinced about the “false positive” effect of performed experiments from which the mentioned results were achieved. In this article, we review the recent literature that provides evidence (from both fronts) of dietary, plant miRNA uptake and functionality in various consumers. Additionally, we discuss possible miRNA transport mechanisms from plant food sources to human cells.
Collapse
Affiliation(s)
- Anna Lukasik
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106 Warsaw, Poland.
| | - Piotr Zielenkiewicz
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106 Warsaw, Poland.
- Department of Plant Molecular Biology, Institute of Experimental Plant Biology and Biotechnology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland.
| |
Collapse
|
635
|
Advancing the use of noncoding RNA in regulatory toxicology: Report of an ECETOC workshop. Regul Toxicol Pharmacol 2016; 82:127-139. [DOI: 10.1016/j.yrtph.2016.09.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 09/19/2016] [Indexed: 12/19/2022]
|
636
|
Gustincich S, Zucchelli S, Mallamaci A. The Yin and Yang of nucleic acid-based therapy in the brain. Prog Neurobiol 2016; 155:194-211. [PMID: 27887908 DOI: 10.1016/j.pneurobio.2016.11.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 11/16/2016] [Accepted: 11/20/2016] [Indexed: 02/06/2023]
Abstract
The post-genomic era has unveiled the existence of a large repertory of non-coding RNAs and repetitive elements that play a fundamental role in cellular homeostasis and dysfunction. These may represent unprecedented opportunities to modify gene expression at the right time in the correct space in vivo, providing an almost unlimited reservoir of new potential pharmacological agents. Hijacking their mode of actions, the druggable genome can be extended to regulatory RNAs and DNA elements in a scalable fashion. Here, we discuss the state-of-the-art of nucleic acid-based drugs to treat neurodegenerative diseases. Beneficial effects can be obtained by inhibiting (Yin) and increasing (Yang) gene expression, depending on the disease and the drug target. Together with the description of the current use of inhibitory RNAs (small inhibitory RNAs and antisense oligonucleotides) in animal models and clinical trials, we discuss the molecular basis and applications of new classes of activatory RNAs at transcriptional (RNAa) and translational (SINEUP) levels.
Collapse
Affiliation(s)
- Stefano Gustincich
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia (IIT), Genova, Italy; Area of Neuroscience, SISSA, Trieste, Italy.
| | - Silvia Zucchelli
- Area of Neuroscience, SISSA, Trieste, Italy; Department of Health Sciences, Universita' del Piemonte Orientale, Novara, Italy
| | | |
Collapse
|
637
|
Magnetic nanoparticle-induced hyperthermia with appropriate payloads: Paul Ehrlich’s “magic (nano)bullet” for cancer theranostics? Cancer Treat Rev 2016; 50:217-227. [DOI: 10.1016/j.ctrv.2016.09.016] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/16/2016] [Accepted: 09/22/2016] [Indexed: 01/11/2023]
|
638
|
Malcolm DW, Sorrells JE, Van Twisk D, Thakar J, Benoit DSW. Evaluating side effects of nanoparticle-mediated siRNA delivery to mesenchymal stem cells using next generation sequencing and enrichment analysis. Bioeng Transl Med 2016; 1:193-206. [PMID: 27981244 PMCID: PMC5125403 DOI: 10.1002/btm2.10035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/06/2016] [Accepted: 09/13/2016] [Indexed: 12/11/2022] Open
Abstract
RNA interference has immense potential to modulate cell functions. However, effective delivery of small interfering RNA (siRNA) while avoiding deleterious side effects has proven challenging. This study investigates both intended and unintended effects of diblock copolymer nanoparticle (NP) delivery of siRNA delivery to human mesenchymal stem cells (hMSC). Specifically, siRNA delivery was investigated at a range of NP‐siRNA:hMSC ratios with a focus on the effects of NP‐siRNA treatment on hMSC functions. Additionally, next generation RNA sequencing (RNAseq) was used with enrichment analysis to observe side effects in hMSC gene expression. Results show NP‐siRNA delivery is negatively correlated with hMSC density. However, higher NP‐siRNA:hMSC ratios increased cytotoxicity and decreased metabolic activity. hMSC proliferation was largely unaffected by NP‐siRNA treatment, except for a threefold reduction in hMSCs seeded at 4,000 cells/cm2. Flow cytometry reveals that apoptosis is a function of NP‐siRNA treatment time and seeding density; ∼14% of the treated hMSCs seeded at 8,000 cells/cm2 were annexin V+‐siRNA+ 24 hr after treatment, while 11% of the treated population was annexin V+‐siRNA−. RNAseq shows that NP‐siRNA treatment results in transcriptomic changes in hMSCs, while pathway analysis shows upregulation of apoptosis signaling and downregulation of metabolism, cell cycle, and DNA replication pathways, as corroborated by apoptosis, metabolism, and proliferation assays. Additionally, multiple innate immune signaling pathways such as toll‐like receptor, RIG‐I‐like receptor, and nuclear factor‐κB signaling pathways are upregulated. Furthermore, and consistent with traditional siRNA immune activation, cytokine–cytokine receptor signaling was also upregulated. Overall, this study provides insight into NP‐siRNA:hMSC ratios that are favorable for siRNA delivery. Moreover, NP‐siRNA delivery results in side effects across the hMSC transcriptome that suggest activation of the innate immunity that could alter MSC functions associated with their therapeutic potential.
Collapse
Affiliation(s)
- Dominic W Malcolm
- Dept. of Biomedical Engineering University of Rochester Rochester NY 14627; Center for Musculoskeletal Research, University of Rochester Rochester NY14642
| | - Janet E Sorrells
- Dept. of Biomedical Engineering University of Rochester Rochester NY 14627
| | - Daniel Van Twisk
- Dept. of Microbiology and Immunology University of Rochester Rochester NY 14627
| | - Juilee Thakar
- Dept. of Microbiology and Immunology University of Rochester Rochester NY 14627; Dept. of Biostatistics and Computational Biology University of Rochester Rochester NY 14642
| | - Danielle S W Benoit
- Dept. of Biomedical Engineering University of Rochester Rochester NY 14627; Center for Musculoskeletal Research, University of Rochester Rochester NY 14642; Dept. of Chemical Engineering University of Rochester Rochester NY 14627
| |
Collapse
|
639
|
Abstract
The discovery of an ever-expanding plethora of coding and non-coding RNAs with nodal and causal roles in the regulation of lung physiology and disease is reinvigorating interest in the clinical utility of the oligonucleotide therapeutic class. This is strongly supported through recent advances in nucleic acids chemistry, synthetic oligonucleotide delivery and viral gene therapy that have succeeded in bringing to market at least three nucleic acid-based drugs. As a consequence, multiple new candidates such as RNA interference modulators, antisense, and splice switching compounds are now progressing through clinical evaluation. Here, manipulation of RNA for the treatment of lung disease is explored, with emphasis on robust pharmacological evidence aligned to the five pillars of drug development: exposure to the appropriate tissue, binding to the desired molecular target, evidence of the expected mode of action, activity in the relevant patient population and commercially viable value proposition.
Collapse
|
640
|
MicroRNA miR-204 and miR-1236 inhibit hepatitis B virus replication via two different mechanisms. Sci Rep 2016; 6:34740. [PMID: 27734898 PMCID: PMC5062086 DOI: 10.1038/srep34740] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 09/19/2016] [Indexed: 12/30/2022] Open
Abstract
Hepatitis B virus (HBV) is a major human pathogen. In this study, we found that miR-204 and miR-1236 were down-regulated in HBV-producing cells, and each could suppress HBV replication. Using a bioinformatic approach and a reporter assay, we identified miR-1236, which can reduce HBV replication and protein production by directly targeting at HBV specific mRNA. In contrast, miR-204 was identified by a microarray approach, and had no effect on HBV RNA and protein production. Surprisingly, miR-204 could inhibit HBV pregenomic RNA encapsidation and capsid assembly. We further demonstrated that HBV suppressed miR-204 expression via activating a host transcription factor STAT3. We established a positive feed-forward loop between HBV, miR-204 and STAT3. Interestingly, miR-204 has been considered as a tumor suppressor in some literature. Since the risk for hepatocellular carcinoma (HCC) is significantly increased in chronic HBV patients, it is possible that chronic suppression of miR-204 by HBV contributes to HCC incidence. Both miR-204 and miR-1236 might be useful for developing new therapeutics against HBV.
Collapse
|
641
|
Patel MR, Kozuch SD, Cultrara CN, Yadav R, Huang S, Samuni U, Koren J, Chiosis G, Sabatino D. RNAi Screening of the Glucose-Regulated Chaperones in Cancer with Self-Assembled siRNA Nanostructures. NANO LETTERS 2016; 16:6099-6108. [PMID: 27669096 PMCID: PMC5378679 DOI: 10.1021/acs.nanolett.6b02274] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
The emerging field of RNA nanotechnology has been used to design well-programmed, self-assembled nanostructures for applications in chemistry, biology, and medicine. At the forefront of its utility in cancer is the unrestricted ability to self-assemble multiple siRNAs within a single nanostructure formulation for the RNAi screening of a wide range of oncogenes while potentiating the gene therapy of malignant tumors. In our RNAi nanotechnology approach, V- and Y-shape RNA templates were designed and constructed for the self-assembly of discrete, higher-ordered siRNA nanostructures targeting the oncogenic glucose regulated chaperones. The GRP78-targeting siRNAs self-assembled into genetically encoded spheres, triangles, squares, pentagons and hexagons of discrete sizes and shapes according to TEM imaging. Furthermore, gel electrophoresis, thermal denaturation, and CD spectroscopy validated the prerequisite siRNA hybrids for their RNAi application. In a 24 sample siRNA screen conducted within the AN3CA endometrial cancer cells known to overexpress oncogenic GRP78 activity, the self-assembled siRNAs targeting multiple sites of GRP78 expression demonstrated more potent and long-lasting anticancer activity relative to their linear controls. Extending the scope of our RNAi screening approach, the self-assembled siRNA hybrids (5 nM) targeting of GRP-75, 78, and 94 resulted in significant (50-95%) knockdown of the glucose regulated chaperones, which led to synergistic effects in tumor cell cycle arrest (50-80%) and death (50-60%) within endometrial (AN3CA), cervical (HeLa), and breast (MDA-MB-231) cancer cell lines. Interestingly, a nontumorigenic lung (MRC5) cell line displaying normal glucose regulated chaperone levels was found to tolerate siRNA treatment and demonstrated less toxicity (5-20%) relative to the cancer cells that were found to be addicted to glucose regulated chaperones. These remarkable self-assembled siRNA nanostructures may thus encompass a new class of potent siRNAs that may be useful in screening important oncogene targets while improving siRNA therapeutic efficacy and specificity in cancer.
Collapse
Affiliation(s)
- Mayurbhai R. Patel
- Program in Chemical Biology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Department of Chemistry and Biochemistry, Seton Hall University, South Orange, New Jersey 07079, United States
| | - Stephen D. Kozuch
- Department of Chemistry and Biochemistry, Seton Hall University, South Orange, New Jersey 07079, United States
| | - Christopher N. Cultrara
- Department of Chemistry and Biochemistry, Seton Hall University, South Orange, New Jersey 07079, United States
| | - Reeta Yadav
- Department of Chemistry and Biochemistry, Queens College, City University of New York, 65-30 Kissena Blvd., Flushing, New York 11367, United States
- Ph.D. Programs in Chemistry and Biochemistry, The Graduate Center of the City University of New York, New York City, New York 10016, United States
| | - Suiying Huang
- Department of Chemistry and Biochemistry, Queens College, City University of New York, 65-30 Kissena Blvd., Flushing, New York 11367, United States
- Ph.D. Programs in Chemistry and Biochemistry, The Graduate Center of the City University of New York, New York City, New York 10016, United States
| | - Uri Samuni
- Department of Chemistry and Biochemistry, Queens College, City University of New York, 65-30 Kissena Blvd., Flushing, New York 11367, United States
- Ph.D. Programs in Chemistry and Biochemistry, The Graduate Center of the City University of New York, New York City, New York 10016, United States
| | - John Koren
- Program in Chemical Biology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Gabriela Chiosis
- Program in Chemical Biology and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - David Sabatino
- Department of Chemistry and Biochemistry, Seton Hall University, South Orange, New Jersey 07079, United States
- Corresponding Author.
| |
Collapse
|
642
|
Tsekoura EK, K C RB, Uludag H. Biomaterials to Facilitate Delivery of RNA Agents in Bone Regeneration and Repair. ACS Biomater Sci Eng 2016; 3:1195-1206. [PMID: 33440509 DOI: 10.1021/acsbiomaterials.6b00387] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Bone healing after traumatic injuries or pathological diseases remains an important worldwide problem. In search of safer and more effective approaches to bone regeneration and repair, RNA-based therapeutic agents, specifically microRNAs (miRNAs) and short interfering RNA (siRNA), are beginning to be actively explored. In this review, we summarize current attempts to employ miRNAs and siRNAs in preclinical models of bone repair. We provide a summary of current limitations when attempting to utilize bioactive nucleic acids for therapeutic purposes and position the unique aspects of RNA reagents for clinical bone repair. Delivery strategies for RNA reagents are emphasized and nonviral carriers (biomaterial-based) employed to deliver such reagents are reviewed. Critical features of biomaterial carriers and various delivery technologies centered around nanoparticulate systems are highlighted. We conclude with the authors' perspectives on the future of the field, outlining main critical issues important to address as RNA reagents are explored for clinical applications.
Collapse
Affiliation(s)
- Eleni K Tsekoura
- Department of Chemical & Materials Engineering, Faculty of Engineering, ‡Department of Biomedical Engineering, Faculty of Medicine & Dentistry, and §Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Remant Bahadur K C
- Department of Chemical & Materials Engineering, Faculty of Engineering, Department of Biomedical Engineering, Faculty of Medicine & Dentistry, and §Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Hasan Uludag
- Department of Chemical & Materials Engineering, Faculty of Engineering, Department of Biomedical Engineering, Faculty of Medicine & Dentistry, and Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| |
Collapse
|
643
|
Andrzejewska W, Pietralik Z, Skupin M, Kozak M. Structural studies of the formation of lipoplexes between siRNA and selected bis-imidazolium gemini surfactants. Colloids Surf B Biointerfaces 2016; 146:598-606. [DOI: 10.1016/j.colsurfb.2016.06.062] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/27/2016] [Accepted: 06/28/2016] [Indexed: 10/21/2022]
|
644
|
Delivery of RNAi Therapeutics to the Airways-From Bench to Bedside. Molecules 2016; 21:molecules21091249. [PMID: 27657028 PMCID: PMC6272875 DOI: 10.3390/molecules21091249] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 09/05/2016] [Accepted: 09/13/2016] [Indexed: 12/12/2022] Open
Abstract
RNA interference (RNAi) is a potent and specific post-transcriptional gene silencing process. Since its discovery, tremendous efforts have been made to translate RNAi technology into therapeutic applications for the treatment of different human diseases including respiratory diseases, by manipulating the expression of disease-associated gene(s). Similar to other nucleic acid-based therapeutics, the major hurdle of RNAi therapy is delivery. Pulmonary delivery is a promising approach of delivering RNAi therapeutics directly to the airways for treating local conditions and minimizing systemic side effects. It is a non-invasive route of administration that is generally well accepted by patients. However, pulmonary drug delivery is a challenge as the lungs pose a series of anatomical, physiological and immunological barriers to drug delivery. Understanding these barriers is essential for the development an effective RNA delivery system. In this review, the different barriers to pulmonary drug delivery are introduced. The potential of RNAi molecules as new class of therapeutics, and the latest preclinical and clinical studies of using RNAi therapeutics in different respiratory conditions are discussed in details. We hope this review can provide some useful insights for moving inhaled RNAi therapeutics from bench to bedside.
Collapse
|
645
|
Green DW, Watson GS, Watson JA, Lee DJ, Lee JM, Jung HS. Diversification and enrichment of clinical biomaterials inspired by Darwinian evolution. Acta Biomater 2016; 42:33-45. [PMID: 27381524 DOI: 10.1016/j.actbio.2016.06.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Revised: 06/11/2016] [Accepted: 06/21/2016] [Indexed: 02/06/2023]
Abstract
UNLABELLED Regenerative medicine and biomaterials design are driven by biomimicry. There is the essential requirement to emulate human cell, tissue, organ and physiological complexity to ensure long-lasting clinical success. Biomimicry projects for biomaterials innovation can be re-invigorated with evolutionary insights and perspectives, since Darwinian evolution is the original dynamic process for biological organisation and complexity. Many existing human inspired regenerative biomaterials (defined as a nature generated, nature derived and nature mimicking structure, produced within a biological system, which can deputise for, or replace human tissues for which it closely matches) are without important elements of biological complexity such as, hierarchy and autonomous actions. It is possible to engineer these essential elements into clinical biomaterials via bioinspired implementation of concepts, processes and mechanisms played out during Darwinian evolution; mechanisms such as, directed, computational, accelerated evolutions and artificial selection contrived in the laboratory. These dynamos for innovation can be used during biomaterials fabrication, but also to choose optimal designs in the regeneration process. Further evolutionary information can help at the design stage; gleaned from the historical evolution of material adaptations compared across phylogenies to changes in their environment and habitats. Taken together, harnessing evolutionary mechanisms and evolutionary pathways, leading to ideal adaptations, will eventually provide a new class of Darwinian and evolutionary biomaterials. This will provide bioengineers with a more diversified and more efficient innovation tool for biomaterial design, synthesis and function than currently achieved with synthetic materials chemistry programmes and rational based materials design approach, which require reasoned logic. It will also inject further creativity, diversity and richness into the biomedical technologies that we make. All of which are based on biological principles. Such evolution-inspired biomaterials have the potential to generate innovative solutions, which match with existing bioengineering problems, in vital areas of clinical materials translation that include tissue engineering, gene delivery, drug delivery, immunity modulation, and scar-less wound healing. STATEMENT OF SIGNIFICANCE Evolution by natural selection is a powerful generator of innovations in molecular, materials and structures. Man has influenced evolution for thousands of years, to create new breeds of farm animals and crop plants, but now molecular and materials can be molded in the same way. Biological molecules and simple structures can be evolved, literally in the laboratory. Furthermore, they are re-designed via lessons learnt from evolutionary history. Through a 3-step process to (1) create variants in material building blocks, (2) screen the variants with beneficial traits/properties and (3) select and support their self-assembly into usable materials, improvements in design and performance can emerge. By introducing biological molecules and small organisms into this process, it is possible to make increasingly diversified, sophisticated and clinically relevant materials for multiple roles in biomedicine.
Collapse
Affiliation(s)
- D W Green
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Republic of Korea; Oral Biosciences, Faculty of Dentistry, The University of Hong Kong, 34, Hospital Road, Hong Kong SAR
| | - G S Watson
- School of Science & Engineering, University of the Sunshine Coast, Sippy Downs, QLD 4556, Australia
| | - J A Watson
- School of Science & Engineering, University of the Sunshine Coast, Sippy Downs, QLD 4556, Australia
| | - D-J Lee
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - J-M Lee
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - H-S Jung
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Republic of Korea; Oral Biosciences, Faculty of Dentistry, The University of Hong Kong, 34, Hospital Road, Hong Kong SAR.
| |
Collapse
|
646
|
Dixon DL, Trankle C, Buckley L, Parod E, Carbone S, Van Tassell BW, Abbate A. A review of PCSK9 inhibition and its effects beyond LDL receptors. J Clin Lipidol 2016; 10:1073-80. [DOI: 10.1016/j.jacl.2016.07.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 07/09/2016] [Indexed: 12/26/2022]
|
647
|
Shih C, Chou SF, Yang CC, Huang JY, Choijilsuren G, Jhou RS. Control and Eradication Strategies of Hepatitis B Virus. Trends Microbiol 2016; 24:739-749. [DOI: 10.1016/j.tim.2016.05.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 05/04/2016] [Accepted: 05/23/2016] [Indexed: 02/07/2023]
|
648
|
Zhu J, Li Z, Wang Q, Liu Y, He J. The contribution of adenines in the catalytic core of 10-23 DNAzyme improved by the 6-amino group modifications. Bioorg Med Chem Lett 2016; 26:4462-4465. [DOI: 10.1016/j.bmcl.2016.07.076] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 07/13/2016] [Accepted: 07/29/2016] [Indexed: 10/21/2022]
|
649
|
Kim HJ, Kim A, Miyata K, Kataoka K. Recent progress in development of siRNA delivery vehicles for cancer therapy. Adv Drug Deliv Rev 2016; 104:61-77. [PMID: 27352638 DOI: 10.1016/j.addr.2016.06.011] [Citation(s) in RCA: 313] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 05/21/2016] [Accepted: 06/16/2016] [Indexed: 12/13/2022]
Abstract
Recent progress in RNA biology has broadened the scope of therapeutic targets of RNA drugs for cancer therapy. However, RNA drugs, typically small interfering RNAs (siRNAs), are rapidly degraded by RNases and filtrated in the kidney, thereby requiring a delivery vehicle for efficient transport to the target cells. To date, various delivery formulations have been developed from cationic lipids, polymers, and/or inorganic nanoparticles for systemic delivery of siRNA to solid tumors. This review describes the current status of clinical trials related to siRNA-based cancer therapy, as well as the remaining issues that need to be overcome to establish a successful therapy. It, then introduces various promising design strategies of delivery vehicles for stable and targeted siRNA delivery, including the prospects for future design.
Collapse
|
650
|
Barata P, Sood AK, Hong DS. RNA-targeted therapeutics in cancer clinical trials: Current status and future directions. Cancer Treat Rev 2016; 50:35-47. [PMID: 27612280 DOI: 10.1016/j.ctrv.2016.08.004] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 08/12/2016] [Indexed: 12/25/2022]
Abstract
Recent advances in RNA delivery and target selection provide unprecedented opportunities for cancer treatment, especially for cancers that are particularly hard to treat with existing drugs. Small interfering RNAs, microRNAs, and antisense oligonucleotides are the most widely used strategies for silencing gene expression. In this review, we summarize how these approaches were used to develop drugs targeting RNA in human cells. Then, we review the current state of clinical trials of these agents for different types of cancer and outcomes from published data. Finally, we discuss lessons learned from completed studies and future directions for this class of drugs.
Collapse
Affiliation(s)
- Pedro Barata
- Department of Solid Tumors, Taussig Cancer Institute, Cleveland Clinic, Cleveland, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|