101
|
Wang Y, Bruggeman KF, Franks S, Gautam V, Hodgetts SI, Harvey AR, Williams RJ, Nisbet DR. Is Viral Vector Gene Delivery More Effective Using Biomaterials? Adv Healthc Mater 2021; 10:e2001238. [PMID: 33191667 DOI: 10.1002/adhm.202001238] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/03/2020] [Indexed: 12/16/2022]
Abstract
Gene delivery has been extensively investigated for introducing foreign genetic material into cells to promote expression of therapeutic proteins or to silence relevant genes. This approach can regulate genetic or epigenetic disorders, offering an attractive alternative to pharmacological therapy or invasive protein delivery options. However, the exciting potential of viral gene therapy has yet to be fully realized, with a number of clinical trials failing to deliver optimal therapeutic outcomes. Reasons for this include difficulty in achieving localized delivery, and subsequently lower efficacy at the target site, as well as poor or inconsistent transduction efficiency. Thus, ongoing efforts are focused on improving local viral delivery and enhancing its efficiency. Recently, biomaterials have been exploited as an option for more controlled, targeted and programmable gene delivery. There is a growing body of literature demonstrating the efficacy of biomaterials and their potential advantages over other delivery strategies. This review explores current limitations of gene delivery and the progress of biomaterial-mediated gene delivery. The combination of biomaterials and gene vectors holds the potential to surmount major challenges, including the uncontrolled release of viral vectors with random delivery duration, poorly localized viral delivery with associated off-target effects, limited viral tropism, and immune safety concerns.
Collapse
Affiliation(s)
- Yi Wang
- Laboratory of Advanced Biomaterials Research School of Engineering The Australian National University Canberra ACT 2601 Australia
| | - Kiara F. Bruggeman
- Laboratory of Advanced Biomaterials Research School of Engineering The Australian National University Canberra ACT 2601 Australia
| | - Stephanie Franks
- Laboratory of Advanced Biomaterials Research School of Engineering The Australian National University Canberra ACT 2601 Australia
| | - Vini Gautam
- Department of Biomedical Engineering The University of Melbourne Melbourne Victoria 3010 Australia
| | - Stuart I. Hodgetts
- School of Human Sciences The University of Western Australia Perth WA 6009 Australia
- Perron Institute for Neurological and Translational Science Perth WA 6009 Australia
| | - Alan R. Harvey
- School of Human Sciences The University of Western Australia Perth WA 6009 Australia
- Perron Institute for Neurological and Translational Science Perth WA 6009 Australia
| | - Richard J. Williams
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT) School of Medicine Deakin University Waurn Ponds VIC 3216 Australia
- Biofab3D St. Vincent's Hospital Fitzroy 3065 Australia
| | - David R. Nisbet
- Laboratory of Advanced Biomaterials Research School of Engineering The Australian National University Canberra ACT 2601 Australia
- Biofab3D St. Vincent's Hospital Fitzroy 3065 Australia
| |
Collapse
|
102
|
Qu J, Li Y, Liao S, Yan J. The Effects of Negative Elements in Environment and Cancer on Female Reproductive System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1300:283-313. [PMID: 33523439 DOI: 10.1007/978-981-33-4187-6_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
With the development of human society, factors that contribute to the impairment of female fertility is accumulating. Lifestyle-related risk factors, occupational risk factors, and iatrogenic factors, including cancer and anti-cancer treatments, have been recognized with their negative effects on the function of female reproductive system. However, the exact influences and their possible mechanism have not been elucidated yet. It is impossible to accurately estimate the indexes of female fertility, but many researchers have put forward that the general fertility has inclined through the past decades. Thus the demand for fertility preservation has increased more and more dramatically. Here we described some of the factors which may influence female reproductive system and methods for fertility preservation in response to female infertility.
Collapse
Affiliation(s)
- Jiangxue Qu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Yuehan Li
- Department of Gynaecology and Obstetrics, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shujie Liao
- Department of Gynaecology and Obstetrics, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Jie Yan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
103
|
Kalita S, Dhayani A, Kumar V, Sujanthi E, Vemula PK. Fate of Biomaterials Post Payload Delivery: Current Understanding and Future Perspectives. NANOTECHNOLOGY IN THE LIFE SCIENCES 2021:141-173. [DOI: 10.1007/978-3-030-61021-0_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
104
|
Jungraithmayr W. Novel Strategies for Endothelial Preservation in Lung Transplant Ischemia-Reperfusion Injury. Front Physiol 2020; 11:581420. [PMID: 33391010 PMCID: PMC7775419 DOI: 10.3389/fphys.2020.581420] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022] Open
Abstract
Lung ischemia reperfusion (IR) injury inevitably occurs during lung transplantation. The pulmonary endothelium is the primary target of IR injury that potentially results in severe pulmonary dysfunction. Over the last decades, various molecules, receptors, and signaling pathways were identified in order to develop treatment strategies for the preservation of the pulmonary endothelium against IR injury. We here review the latest and most promising therapeutic strategies for the protection of the endothelium against IR injury. These include the stabilization of the endothelial glycocalyx, inhibition of endothelial autophagy, inhibition of adhesion molecules, targeting of angiotensin-converting enzyme, and traditional viral and novel non-viral gene transfer approaches. Though some of these strategies proved to be promising in experimental studies, very few of these treatment concepts made the transfer into clinical application. This dilemma underscores the need for more experimental evidence for the translation into clinical studies to invent therapeutic concepts against IR injury-mediated endothelial damage.
Collapse
Affiliation(s)
- Wolfgang Jungraithmayr
- Department of Thoracic Surgery, University Hospital Freiburg, Freiburg, Germany.,Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland.,Department of Thoracic Surgery, University Hospital Rostock, Rostock, Germany
| |
Collapse
|
105
|
Lee C, Choi M, MacKay JA. Live long and active: Polypeptide-mediated assembly of antibody variable fragments. Adv Drug Deliv Rev 2020; 167:1-18. [PMID: 33129938 DOI: 10.1016/j.addr.2020.10.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 10/20/2020] [Accepted: 10/23/2020] [Indexed: 12/17/2022]
Abstract
Antibodies possess multiple biologically relevant features that have been engineered into new therapeutic formats. Two examples include the adaptable specificity of their variable (Fv) region and the extension of plasma circulation times through their crystallizable (Fc) region. Since the invention of the single chain variable fragment (scFv) in 1988, antibody variable regions have been re-engineered into a wide variety of multifunctional nanostructures. Among these strategies, peptide-mediated self-assembly of variable regions through heterologous expression has become a powerful method to produce homogenous, functional biomaterials. This manuscript reviews recent reports of antibody fragments assembled through fusion with peptides and proteins, including elastin-like polypeptides (ELPs), collagen-like polypeptides (CLPs), albumin, transmembrane proteins, leucine zippers, silk protein, and viruses. This review further discusses the current clinical status of engineered antibody fragments and challenges to overcome.
Collapse
Affiliation(s)
- Changrim Lee
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - Minchang Choi
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - J Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA; Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA; Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, United States.
| |
Collapse
|
106
|
Zhang R, Tang L, Tian Y, Ji X, Hu Q, Zhou B, Ding Z, Xu H, Yang L. DP7-C-modified liposomes enhance immune responses and the antitumor effect of a neoantigen-based mRNA vaccine. J Control Release 2020; 328:210-221. [PMID: 32860927 DOI: 10.1016/j.jconrel.2020.08.023] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 08/06/2020] [Accepted: 08/17/2020] [Indexed: 02/08/2023]
Abstract
To date, many clinical trials have been carried out with neoantigen-specific mRNA vaccines, and positive results have been achieved. However, further improvements in the efficiency of the intracellular delivery of mRNA and the production of a stronger immune response are still worth studying. In this study, we used the cholesterol-modified cationic peptide DP7 (VQWRIRVAVIRK), which was developed in our previous study, with a transmembrane structure and immunoadjuvant function to modify DOTAP liposomes to create a common mRNA delivery system. This system was intended to improve the efficiency of the delivery of mRNA encoding individualized neoantigens to dendritic cells (DCs) and enhance the activation of DCs. The system serves dual functions as a carrier and as an immunoadjuvant. As a carrier of mRNA, DP7-C-modified DOTAP liposomes (DOTAP/DP7-C) could transfer mRNA efficiently into different type of DCs in vitro. As an immunoadjuvant, DOTAP/DP7-C liposomes were shown to be more efficacious in stimulating DC maturation, CD103+ DC (contributing to antigen presentation) production and proinflammatory cytokine secretion than DOTAP liposomes both in vitro and in vivo. In animal studies, the subcutaneous administration of DOTAP/DP7-C/LL2 neoantigen-encoding mRNA complexes significantly inhibited the growth of LL2 in situ and the growth of subcutaneous tumors and stimulated the production of antigen-specific lymphocyte reactions, which were superior to the DOTAP/LL2 neoantigen-encoding mRNA complex group. In conclusion, DOTAP/DP7-C liposomes may serve as a potential universal mRNA delivery system, providing a simple method to increase the efficiency of intracellular mRNA delivery and the immunostimulatory activity of DCs.
Collapse
Affiliation(s)
- Rui Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Lin Tang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Yaomei Tian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Xiao Ji
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Qiuyue Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Bailing Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Zhenyu Ding
- Department of Pharmacy and Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, People's Republic of China
| | - Heng Xu
- Precision Medicine Center, State Key Laboratory of Biotherapy, and Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| |
Collapse
|
107
|
Bodbin SE, Denning C, Mosqueira D. Transfection of hPSC-Cardiomyocytes Using Viafect™ Transfection Reagent. Methods Protoc 2020; 3:E57. [PMID: 32784848 PMCID: PMC7564709 DOI: 10.3390/mps3030057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 02/06/2023] Open
Abstract
Twenty years since their first derivation, human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have shown promise in disease modelling research, while their potential for cardiac repair is being investigated. However, low transfection efficiency is a barrier to wider realisation of the potential this model system has to offer. We endeavoured to produce a protocol for improved transfection of hPSC-CMs using the ViafectTM reagent by Promega. Through optimisation of four essential parameters: (i) serum supplementation, (ii) time between replating and transfection, (iii) reagent to DNA ratio and (iv) cell density, we were able to successfully transfect hPSC-CMs to ~95% efficiencies. Transfected hPSC-CMs retained high purity and structural integrity despite a mild reduction in viability, and preserved compatibility with phenotyping assays of hypertrophy. This protocol greatly adds value to the field by overcoming limited transfection efficiencies of hPSC-CMs in a simple and quick approach that ensures sustained expression of transfected genes for at least 14 days, opening new opportunities in mechanistic discovery for cardiac-related diseases.
Collapse
Affiliation(s)
- Sara E. Bodbin
- Division of Cancer & Stem Cells, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Chris Denning
- Division of Cancer & Stem Cells, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Diogo Mosqueira
- Division of Cancer & Stem Cells, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| |
Collapse
|
108
|
Hu LZ, Wang W, Xu JL, Jia YY, Huan ML, Li C, Zhou SY, Zhang BL. Polyethylenimine-based nanovector grafted with mannitol moieties to achieve effective gene delivery and transfection. NANOTECHNOLOGY 2020; 31:325101. [PMID: 32325436 DOI: 10.1088/1361-6528/ab8c76] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Polyethylenimine (PEI), a kind of cationic non-viral gene delivery vector, is capable of stable and efficient transgene expression for gene delivery. However, low transfection efficiency in vivo along with high toxicity limited the further application of gene therapy in the clinic. To enhance gene transfection performance and reduce cytotoxicity of polyethylenimine, branched polyethylenimine-derived cationic polymers BPEI25 k-man-S/L/M/H with different grafting degree with mannitol moieties were prepared and the transfection efficiency was evaluated. Among them, BPEI25 k-man-L showed the best transfection efficiency, lower toxicity, and significantly enhanced long-term systemic transgene expression for 96 h in vivo even at a single-dose administration. The results of cellular uptake mechanism and western-blot experiments revealed that the mannitol modification of BPEI25 k induced and up-regulated the phosphorylation of caveolin-1 and thus enhanced the caveolae-mediated cellular uptake. This class of gene delivery system highlights a paradigmatic approach for the development of novel and safe non-viral vectors for gene therapy.
Collapse
Affiliation(s)
- Li-Zhong Hu
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, People's Republic of China. Department of Pharmacy, Luoyang Polytechnic, Luoyang 471000, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
109
|
Chen W, Hu Y, Ju D. Gene therapy for neurodegenerative disorders: advances, insights and prospects. Acta Pharm Sin B 2020; 10:1347-1359. [PMID: 32963936 PMCID: PMC7488363 DOI: 10.1016/j.apsb.2020.01.015] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/09/2019] [Accepted: 12/06/2019] [Indexed: 02/07/2023] Open
Abstract
Gene therapy is rapidly emerging as a powerful therapeutic strategy for a wide range of neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD). Some early clinical trials have failed to achieve satisfactory therapeutic effects. Efforts to enhance effectiveness are now concentrating on three major fields: identification of new vectors, novel therapeutic targets, and reliable of delivery routes for transgenes. These approaches are being assessed closely in preclinical and clinical trials, which may ultimately provide powerful treatments for patients. Here, we discuss advances and challenges of gene therapy for neurodegenerative disorders, highlighting promising technologies, targets, and future prospects.
Collapse
Key Words
- AADC, aromatic-l-amino-acid
- AAVs, adeno-associated viruses
- AD, Alzheimer's disease
- ARSA, arylsulfatase A
- ASOs, antisense oligonucleotides
- ASPA, aspartoacylase
- Adeno-associated viruses
- Adv, adenovirus
- BBB, blood–brain barrier
- BCSFB, blood–cerebrospinal fluid barrier
- BRB, blood–retina barrier
- Bip, glucose regulated protein 78
- CHOP, CCAAT/enhancer binding homologous protein
- CLN6, ceroidlipofuscinosis neuronal protein 6
- CNS, central nervous system
- CSF, cerebrospinal fluid
- Central nervous system
- Delivery routes
- ER, endoplasmic reticulum
- FDA, U.S. Food and Drug Administration
- GAA, lysosomal acid α-glucosidase
- GAD, glutamic acid decarboxylase
- GDNF, glial derived neurotrophic factor
- Gene therapy
- HD, Huntington's disease
- HSPGs, heparin sulfate proteoglycans
- HTT, mutant huntingtin
- IDS, iduronate 2-sulfatase
- LVs, retrovirus/lentivirus
- Lamp2a, lysosomal-associated membrane protein 2a
- NGF, nerve growth factor
- Neurodegenerative disorders
- PD, Parkinson's disease
- PGRN, Progranulin
- PINK1, putative kinase 1
- PTEN, phosphatase and tensin homolog
- RGCs, retinal ganglion cells
- RNAi, RNA interference
- RPE, retinal pigmented epithelial
- SGSH, lysosomal heparan-N-sulfamidase gene
- SMN, survival motor neuron
- SOD, superoxide dismutase
- SUMF, sulfatase-modifying factor
- TFEB, transcription factor EB
- TPP1, tripeptidyl peptidase 1
- TREM2, triggering receptor expressed on myeloid cells 2
- UPR, unfolded protein response
- ZFPs, zinc finger proteins
- mTOR, mammalian target of rapamycin
- siRNA, small interfering RNA
Collapse
Affiliation(s)
- Wei Chen
- Department of Biological Medicines, Fudan University School of Pharmacy, Shanghai 201203, China
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Yang Hu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Dianwen Ju
- Department of Biological Medicines, Fudan University School of Pharmacy, Shanghai 201203, China
| |
Collapse
|
110
|
Xing Q, Pan Y, Hu Y, Wang L. Review of the Biomolecular Modification of the Metal-Organ-Framework. Front Chem 2020; 8:642. [PMID: 32850658 PMCID: PMC7399348 DOI: 10.3389/fchem.2020.00642] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/22/2020] [Indexed: 12/24/2022] Open
Abstract
Metal-organ frameworks (MOFs), as a kind of novel artificial material, have been widely studied in the field of chemistry. MOFs are capable of high loading capacities, controlled release, plasticity, and biosafety because of their porous structure and have been gradually functionalized as a drug carrier. Recently, a completely new strategy of combining biomolecules, such as oligonucleotides, polypeptides, and nucleic acids, with MOF nanoparticles was proposed. The synthetic bio-MOFs conferred strong protection and endowed the MOFs with particular biological functions. Biomolecular modification of MOFs to form bridges for communication between different subjects has received increased attention. This review will focus on bio-MOFs modification methods and discuss the advantages, applications, prospects, and challenges of using MOFs in the field of biomolecule delivery.
Collapse
Affiliation(s)
| | | | | | - Long Wang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
111
|
Zhang J, Li Y, Li J, Shi Y, Hu J, Yang G. Surfce Functionalized via AdLAMA3 Multilayer Coating for Re-epithelization Around Titanium Implants. Front Bioeng Biotechnol 2020; 8:624. [PMID: 32596232 PMCID: PMC7300264 DOI: 10.3389/fbioe.2020.00624] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/21/2020] [Indexed: 11/29/2022] Open
Abstract
The peri-implant epithelium (PIE) forms a crucial seal between the oral environment and the implant surface. Compared with the junctional epithelium (JE), the biological sealing of PIE is fragile, which lacks hemidesmosomes (HDs) and internal basal lamina (extracellular matrix containing laminin332, IBL) on the upper part of the interface. In the study, we aim to prepare a coating with good biocompatibility and ability to immobilize the recombinant adenovirus vector of LAMA3 (AdLAMA3) for promoting the re-epithelization of PIE. The titanium surface functionalized with AdLAMA3 was established via layer-by-layer assembly technique and antibody-antigen specific binding. The biological evaluations including cell adhesion and the re-epithelization of PIE were investigated. The results in vitro demonstrated that the AdLAMA3 coating could improve epithelial cell attachment and cell spreading in the early stage. In vivo experiments indicated that the AdLAMA3 coating on the implant surface has the potential to accelerate the healing of the PIE, and could promote the expression of laminin α3 and the formation of hemidesmosomes. This study might provide a novel approach and experimental evidence for the precise attachment of LAMA3 to titanium surfaces. The process could improve the re-epithelization of PIE.
Collapse
Affiliation(s)
- Jing Zhang
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Yongzheng Li
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Jialu Li
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Yuan Shi
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Jinxing Hu
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Guoli Yang
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| |
Collapse
|
112
|
Kletzmayr A, Ivarsson ME, Leroux JC. Investigational Therapies for Primary Hyperoxaluria. Bioconjug Chem 2020; 31:1696-1707. [PMID: 32539351 DOI: 10.1021/acs.bioconjchem.0c00268] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recent years have brought exciting new insights in the field of primary hyperoxaluria (PH), both on a basic research level as well as through the progress of novel therapeutics in clinical development. To date, very few supportive measures are available for patients suffering from PH, which, together with the severity of the disorder, make disease management challenging. Basic and clinical research and development efforts range from correcting the underlying gene mutations, preventing calcium oxalate crystal-induced kidney damage, to the administration of probiotics favoring the intestinal secretion of excess oxalate. In this review, current advances in the development of those strategies are presented and discussed.
Collapse
Affiliation(s)
- Anna Kletzmayr
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | | | - Jean-Christophe Leroux
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
113
|
Zhang Y, Liu Z. Oncolytic Virotherapy for Malignant Tumor: Current Clinical Status. Curr Pharm Des 2020; 25:4251-4263. [PMID: 31682207 DOI: 10.2174/1381612825666191104090544] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 10/29/2019] [Indexed: 12/12/2022]
Abstract
Oncolytic viruses, as novel biological anti-tumor agents, provide anti-tumor therapeutic effects by different mechanisms including directly selective tumor cell lysis and secondary systemic anti-tumor immune responses. Some wide-type and genetically engineered oncolytic viruses have been applied in clinical trials. Among them, T-Vec has a significant therapeutic effect on melanoma patients and received the approval of the US Food and Drug Administration (FDA) as the first oncolytic virus to treat cancer in the US. However, the mechanisms of virus interaction with tumor and immune systems have not been clearly elucidated and there are still no "gold standards" for instructions of virotherapy in clinical trials. This Review collected the recent clinical trials data from 2005 to summarize the basic oncolytic viruses biology, describe the application in recent clinical trials, and discuss the challenges in the application of oncolytic viruses in clinical trials.
Collapse
Affiliation(s)
- Yuhui Zhang
- Department of Spine Surgery, Renji Hospital, Medical School, Shanghai Jiaotong University, Shanghai, China
| | - Zhuoming Liu
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, United States
| |
Collapse
|
114
|
Buck TM, Wijnholds J. Recombinant Adeno-Associated Viral Vectors (rAAV)-Vector Elements in Ocular Gene Therapy Clinical Trials and Transgene Expression and Bioactivity Assays. Int J Mol Sci 2020; 21:E4197. [PMID: 32545533 PMCID: PMC7352801 DOI: 10.3390/ijms21124197] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 02/06/2023] Open
Abstract
Inherited retinal dystrophies and optic neuropathies cause chronic disabling loss of visual function. The development of recombinant adeno-associated viral vectors (rAAV) gene therapies in all disease fields have been promising, but the translation to the clinic has been slow. The safety and efficacy profiles of rAAV are linked to the dose of applied vectors. DNA changes in the rAAV gene cassette affect potency, the expression pattern (cell-specificity), and the production yield. Here, we present a library of rAAV vectors and elements that provide a workflow to design novel vectors. We first performed a meta-analysis on recombinant rAAV elements in clinical trials (2007-2020) for ocular gene therapies. We analyzed 33 unique rAAV gene cassettes used in 57 ocular clinical trials. The rAAV gene therapy vectors used six unique capsid variants, 16 different promoters, and six unique polyadenylation sequences. Further, we compiled a list of promoters, enhancers, and other sequences used in current rAAV gene cassettes in preclinical studies. Then, we give an update on pro-viral plasmid backbones used to produce the gene therapy vectors, inverted terminal repeats, production yield, and rAAV safety considerations. Finally, we assess rAAV transgene and bioactivity assays applied to cells or organoids in vitro, explants ex vivo, and clinical studies.
Collapse
Affiliation(s)
- Thilo M. Buck
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZC Leiden, The Netherlands;
| | - Jan Wijnholds
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZC Leiden, The Netherlands;
- Netherlands Institute of Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW), 1105 BA Amsterdam, The Netherlands
| |
Collapse
|
115
|
He Y, Wang M, Li X, Yu T, Gao X. Targeted MIP-3β plasmid nanoparticles induce dendritic cell maturation and inhibit M2 macrophage polarisation to suppress cancer growth. Biomaterials 2020; 249:120046. [PMID: 32325346 DOI: 10.1016/j.biomaterials.2020.120046] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/05/2020] [Accepted: 04/09/2020] [Indexed: 02/05/2023]
Abstract
In recent decades, cancer immunotherapy has demonstrated considerable clinical advantages in cancer therapy. Particularly, the use of immunological gene therapy continues to grow in this field. Macrophage Inflammatory Protein 3 Beta (MIP-3β) has emerged as a potential immunomodulator for anti-cancer treatments by enhancing the interaction among immune responses. In this study, we demonstrate an innovative targeted gene delivery system based on a self-assembly technique with 1,2-Dioleoyl-3-trimethylammonium-propane (DOTAP), Methoxy poly(ethylene glycol)-poly(lactide) (MPEG-PLA), and folic acid modified poly(ethylene glycol)-poly(ε-caprolactone) (FA-PEG-PCL) (FDMCA). Results showed that the expression of MIP-3β was up-regulated in cancer cells following the transfection with FDMCA-pMIP-3β, in comparison with cells transfected with DMCA-pMIP-3β. The supernatants collected from cancer cells transfected with FDMCA-pMIP-3β and DMCA-pMIP-3β both instigate dendritic cell maturation, M1 polarisation of macrophages, activation and presentation of cytotoxicity in lymphocytes. Moreover, tumor growth and metastasis were markedly inhibited following the administration of the FDMCA-pMIP-3β complex in both subcutaneous and pulmonary metastasis mice models, which is attributed to reduced angiogenesis, enhanced cancer cell apoptosis, and suppressed proliferation by activation of the immune system. Our study suggests that the MIP-3β plasmid and FDMCA complex provide a new approach for the treatment of breast cancer.
Collapse
Affiliation(s)
- Yihong He
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, PR China
| | - Manni Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, PR China
| | - Xiaoling Li
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, PR China
| | - Ting Yu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, PR China; Department of Pathology and Laboratory of Pathology, State Key Laboratory of Biotherapy, West China Hospital, West China School of Medicine, Sichuan University, PR China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, PR China.
| |
Collapse
|
116
|
Zhou J, Kroll AV, Holay M, Fang RH, Zhang L. Biomimetic Nanotechnology toward Personalized Vaccines. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1901255. [PMID: 31206841 PMCID: PMC6918015 DOI: 10.1002/adma.201901255] [Citation(s) in RCA: 197] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 04/07/2019] [Indexed: 04/14/2023]
Abstract
While traditional approaches for disease management in the era of modern medicine have saved countless lives and enhanced patient well-being, it is clear that there is significant room to improve upon the current status quo. For infectious diseases, the steady rise of antibiotic resistance has resulted in super pathogens that do not respond to most approved drugs. In the field of cancer treatment, the idea of a cure-all silver bullet has long been abandoned. As a result of the challenges facing current treatment and prevention paradigms in the clinic, there is an increasing push for personalized therapeutics, where plans for medical care are established on a patient-by-patient basis. Along these lines, vaccines, both against bacteria and tumors, are a clinical modality that could benefit significantly from personalization. Effective vaccination strategies could help to address many challenging disease conditions, but current vaccines are limited by factors such as a lack of potency and antigenic breadth. Recently, researchers have turned toward the use of biomimetic nanotechnology as a means of addressing these hurdles. Recent progress in the development of biomimetic nanovaccines for antibacterial and anticancer applications is discussed, with an emphasis on their potential for personalized medicine.
Collapse
Affiliation(s)
- Jiarong Zhou
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ashley V Kroll
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Maya Holay
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ronnie H Fang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
117
|
Shen H, Huang X, Min J, Le S, Wang Q, Wang X, Dogan AA, Liu X, Zhang P, Draz MS, Xiao J. Nanoparticle Delivery Systems for DNA/RNA and their Potential Applications in Nanomedicine. Curr Top Med Chem 2020; 19:2507-2523. [PMID: 31775591 DOI: 10.2174/1568026619666191024170212] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 10/06/2019] [Accepted: 10/07/2019] [Indexed: 02/04/2023]
Abstract
The rapid development of nanotechnology has a great influence on the fields of biology, physiology, and medicine. Over recent years, nanoparticles have been widely presented as nanocarriers to help the delivery of gene, drugs, and other therapeutic agents with cellular targeting ability. Advances in the understanding of gene delivery and RNA interference (RNAi)-based therapy have brought increasing attention to understanding and tackling complex genetically related diseases, such as cancer, cardiovascular and pulmonary diseases, autoimmune diseases and infections. The combination of nanocarriers and DNA/RNA delivery may potentially improve their safety and therapeutic efficacy. However, there still exist many challenges before this approach can be practiced in the clinic. In this review, we provide a comprehensive summary on the types of nanoparticle systems used as nanocarriers, highlight the current use of nanocarriers in recombinant DNA and RNAi molecules delivery, and the current landscape of gene-based nanomedicine-ranging from diagnosis to therapeutics. Finally, we briefly discuss the biosafety concerns and limitations in the preclinical and clinical development of nanoparticle gene systems.
Collapse
Affiliation(s)
- Hua Shen
- Department of Cardiothoracic Surgery, Changzheng Hospital, Second Military Medical University, Fengyang Road 415#, Shanghai 200003, China.,Department of Cardiovascular Surgery, Institute of Cardiac Surgery, PLA General Hospital, Beijing, China
| | - Xiaoyi Huang
- Department of Pathology, Changhai Hospital, Second Military Medical University, Changhai Road 168#, Shanghai 200433, China
| | - Jie Min
- Department of Cardiothoracic Surgery, Bethune International Peace Hospital, Shijiazhuang, China
| | - Shiguan Le
- Department of Cardiothoracic Surgery, Changzheng Hospital, Second Military Medical University, Fengyang Road 415#, Shanghai 200003, China
| | - Qing Wang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Second Military Medical University, Fengyang Road 415#, Shanghai 200003, China
| | - Xi Wang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Second Military Medical University, Fengyang Road 415#, Shanghai 200003, China
| | - Asli Aybike Dogan
- Department of Bioengineering, Graduate School of Natural and Applied Sciences, Ege University, 35100 Bornova-Izmir, Turkey
| | - Xiangsheng Liu
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, United States
| | - Pengfei Zhang
- Department of Central Laboratory, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Mohamed S Draz
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139, United States.,Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford St., Cambridge, MA 02138, United States.,Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Jian Xiao
- Department of Cardiothoracic Surgery, Changzheng Hospital, Second Military Medical University, Fengyang Road 415#, Shanghai 200003, China
| |
Collapse
|
118
|
|
119
|
Efficient and Highly Specific Gene Transfer Using Mutated Lentiviral Vectors Redirected with Bispecific Antibodies. mBio 2020; 11:mBio.02990-19. [PMID: 31964730 PMCID: PMC6989108 DOI: 10.1128/mbio.02990-19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The goal of gene therapy is specific delivery and expression of therapeutic genes to target cells and tissues. Common lentivirus (LV) vectors are efficient gene delivery vehicles but offer little specificity. Here, we report an effective and versatile strategy to redirect LV to target cells using bispecific antibodies (bsAbs) that bind both cell receptors and LV envelope domains. Importantly, we ablated the native receptor binding of LV to minimize off-target transduction. Coupling bsAb specificity and ablated native LV tropism synergistically enhanced the selectivity of our targeted gene delivery system. The modular nature of our bsAb-based redirection enables facile targeting of the same LV to diverse tissues/cells. By abrogating the native broad tropism of LV, our bsAb-LV redirection strategy may enable lentivirus-based gene delivery in vivo, expanding the current use of LV beyond ex vivo applications. Despite their exceptional potencies, the broad tropism of most commonly used lentivirus (LV) vectors limits their use for targeted gene delivery in vivo. We hypothesized that we could improve the specificity of LV targeting by coupling (i) reduction of their binding to off-target cells with (ii) redirection of the vectors with a bispecific antibody (bsAb) that binds both LV and receptors on target cells. As a proof of concept, we pseudotyped nonreplicating LV using a mutated Sindbis envelope (mSindbis) with ablated binding to native receptors, while retaining the capacity to facilitate efficient fusion and endosomal escape. We then evaluated the transduction potencies of the mSindbis LV for HER2-positive (HER2+) (SKBR3) breast and HER2-negative (HER2−) (A2780) cells when redirected with different bsAbs. mSindbis LV alone failed to induce appreciable green fluorescent protein (GFP) expression in either cell. When mixed with HER2-targeting bsAb, mSindbis LV was exceptionally potent, transducing 12% to 16% of the SKBR3 cells at a multiplicity of infection (MOI [ratio of viral genome copies to target cells]) of 3. Transduction was highly specific, resulting in ∼50-fold-greater selectivity toward SKBR3 cells versus A2780 cells. Redirecting mSindbis LV led to a 10-fold improvement in cell-specific targeting compared to redirecting wild-type Sindbis LV with the same bsAb, underscoring the importance of ablating native virus tropism in order to maximize targeting specificity. The redirection of mutated LV using bsAb represents a potent and highly versatile platform for targeted gene therapy.
Collapse
|
120
|
Yang TC, Chang CY, Yarmishyn AA, Mao YS, Yang YP, Wang ML, Hsu CC, Yang HY, Hwang DK, Chen SJ, Tsai ML, Lai YH, Tzeng Y, Chang CC, Chiou SH. Carboxylated nanodiamond-mediated CRISPR-Cas9 delivery of human retinoschisis mutation into human iPSCs and mouse retina. Acta Biomater 2020; 101:484-494. [PMID: 31672582 DOI: 10.1016/j.actbio.2019.10.037] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 10/14/2019] [Accepted: 10/24/2019] [Indexed: 01/08/2023]
Abstract
Nanodiamonds (NDs) are considered to be relatively safe carbon nanomaterials used for the transmission of DNA, proteins and drugs. The feasibility of utilizing the NDs to deliver CRISPR-Cas9 system for gene editing has not been clearly studied. Therefore, in this study, we aimed to use NDs as the carriers of CRISPR-Cas9 components designed to introduce the mutation in RS1 gene associated with X-linked retinoschisis (XLRS). ND particles with a diameter of 3 nm were functionalized by carboxylation of the surface and covalently conjugated with fluorescent mCherry protein. Two linear DNA constructs were attached to the conjugated mCherry: one encoded Cas9 endonuclease and GFP reporter, another encoded sgRNA and contained insert of HDR template designed to introduce RS1 c.625C>T mutation. Such nanoparticles were successfully delivered and internalized by human iPSCs and mouse retinas, the efficiency of internalization was significantly improved by mixing with BSA. The delivery of ND particles led to introduction of RS1 c.625C>T mutation in both human iPSCs and mouse retinas. Rs1 gene editing in mouse retinas resulted in several pathological features typical for XLRS, such as aberrant photoreceptor structure. To conclude, our ND-based CRISPR-Cas9 delivery system can be utilized as a tool for creating in vitro and in vivo disease models of XLRS. STATEMENT OF SIGNIFICANCE: X-linked retinoschisis (XLRS) is a prevalent hereditary retinal disease, which is caused by mutations in RS1 gene, whose product is important for structural organization of the retina. The recent development of genome editing techniques such as CRISPR-Cas9 significantly improved the prospects for better understanding the pathology and development of treatment for this disease. Firstly, gene editing can allow development of appropriate in vitro and in vivo disease models; secondly, CRISPR-Cas9 can be applied for gene therapy by removing the disease-causative mutation in vivo. The major prerequisite for these approaches is to develop safe and efficient CRISPR-Cas9 delivery system. In this study, we tested specifically modified nanodiamonds for such a delivery system. We were able to introduce Rs1 mutation into the mouse retina and, importantly, observed several XLRS-specific effects.
Collapse
|
121
|
Zhou Y, Han S, Liang Z, Zhao M, Liu G, Wu J. Progress in arginine-based gene delivery systems. J Mater Chem B 2020; 8:5564-5577. [DOI: 10.1039/d0tb00498g] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Arginine based gene delivery systems with enhanced membrane penetration and lower cytotoxicity greatly enrich the gene vectors library and outline a new development direction of gene delivery.
Collapse
Affiliation(s)
- Yang Zhou
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province
- School of Biomedical Engineering
- Sun Yat-sen University
- Guangzhou
- China
| | - Shuyan Han
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province
- School of Biomedical Engineering
- Sun Yat-sen University
- Guangzhou
- China
| | - Zhiqing Liang
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province
- School of Biomedical Engineering
- Sun Yat-sen University
- Guangzhou
- China
| | - Meng Zhao
- Shenzhen Lansi Institute of Artificial Intelligence in Medicine
- Shenzhen
- China
| | - Guiting Liu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province
- School of Biomedical Engineering
- Sun Yat-sen University
- Guangzhou
- China
| | - Jun Wu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province
- School of Biomedical Engineering
- Sun Yat-sen University
- Guangzhou
- China
| |
Collapse
|
122
|
Kim SH, Lee S, Lee H, Cho M, Schaffer DV, Jang JH. AAVR-Displaying Interfaces: Serotype-Independent Adeno-Associated Virus Capture and Local Delivery Systems. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:432-443. [PMID: 31670142 PMCID: PMC6831863 DOI: 10.1016/j.omtn.2019.09.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/17/2019] [Accepted: 09/17/2019] [Indexed: 12/12/2022]
Abstract
Interfacing gene delivery vehicles with biomaterials has the potential to play a key role in diversifying gene transfer capabilities, including localized, patterned, and controlled delivery. However, strategies for modifying biomaterials to interact with delivery vectors must be redesigned whenever new delivery vehicles and applications are explored. We have developed a vector-independent biomaterial platform capable of interacting with various adeno-associated viral (AAV) serotypes. A water-soluble, cysteine-tagged, recombinant protein version of the recently discovered multi-AAV serotype receptor (AAVR), referred to as cys-AAVR, was conjugated to maleimide-displaying polycaprolactone (PCL) materials using click chemistry. The resulting cys-AAVR-PCL system bound to a broad range of therapeutically relevant AAV serotypes, thereby providing a platform capable of modulating the delivery of all AAV serotypes. Intramuscular injection of cys-AAVR-PCL microspheres with bound AAV vectors resulted in localized and sustained gene delivery as well as reduced spread to off-target organs compared to a vector solution. This cys-AAVR-PCL system is thus an effective approach for biomaterial-based AAV gene delivery for a broad range of therapeutic applications.
Collapse
Affiliation(s)
- Seung-Hyun Kim
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, Korea
| | - Slgirim Lee
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, Korea; Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA
| | - Heehyung Lee
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, Korea
| | - Mira Cho
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, Korea
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720-3220, USA; Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720-3220, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720-3220, USA.
| | - Jae-Hyung Jang
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, Korea.
| |
Collapse
|
123
|
Shao YT, Ma L, Zhang TH, Xu TR, Ye YC, Liu Y. The Application of the RNA Interference Technologies for KRAS: Current Status, Future Perspective and Associated Challenges. Curr Top Med Chem 2019; 19:2143-2157. [PMID: 31456522 DOI: 10.2174/1568026619666190828162217] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/26/2019] [Accepted: 07/07/2019] [Indexed: 02/07/2023]
Abstract
KRAS is a member of the murine sarcoma virus oncogene-RAS gene family. It plays an important role in the prevention, diagnosis and treatment of tumors during tumor cell growth and angiogenesis. KRAS is the most commonly mutated oncogene in human cancers, such as pancreatic cancers, colon cancers, and lung cancers. Detection of KRAS gene mutation is an important indicator for tracking the status of oncogenes, highlighting the developmental prognosis of various cancers, and the efficacy of radiotherapy and chemotherapy. However, the efficacy of different patients in clinical treatment is not the same. Since RNA interference (RNAi) technologies can specifically eliminate the expression of specific genes, these technologies have been widely used in the field of gene therapy for exploring gene function, infectious diseases and malignant tumors. RNAi refers to the phenomenon of highly specific degradation of homologous mRNA induced by double-stranded RNA (dsRNA), which is highly conserved during evolution. There are three classical RNAi technologies, including siRNA, shRNA and CRISPR-Cas9 system, and a novel synthetic lethal interaction that selectively targets KRAS mutant cancers. Therefore, the implementation of individualized targeted drug therapy has become the best choice for doctors and patients. Thus, this review focuses on the current status, future perspective and associated challenges in silencing of KRAS with RNAi technology.
Collapse
Affiliation(s)
- Yu-Ting Shao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Li Ma
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Tie-Hui Zhang
- The First People's Hospital of Heishan County, Jinzhou city, Liaoning, Jinzhou 121400, China
| | - Tian-Rui Xu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Yuan-Chao Ye
- Department of Internal Medicine, Gastroenterology and Hepatology, University of Iowa, Iowa City, IA 52242, United States.,Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, United States
| | - Ying Liu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| |
Collapse
|
124
|
Yan X, Li S, Qu Y, Wang W, Chen B, Liu S, Ma X, Yu X. Redox-Responsive Multifunctional Polypeptides Conjugated with Au Nanoparticles for Tumor-Targeting Gene Therapy and Their 1 + 1 > 2 Synergistic Effects. ACS Biomater Sci Eng 2019; 6:463-473. [DOI: 10.1021/acsbiomaterials.9b01581] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Xinxin Yan
- Laboratory of Polymer Composites Engineering, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Shengran Li
- Laboratory of Polymer Composites Engineering, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Yangchun Qu
- Department of Radiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Wenliang Wang
- Laboratory of Polymer Composites Engineering, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Binggang Chen
- Laboratory of Polymer Composites Engineering, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Sanrong Liu
- Laboratory of Polymer Composites Engineering, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Xiaojing Ma
- Laboratory of Polymer Composites Engineering, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Xifei Yu
- Laboratory of Polymer Composites Engineering, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- University of Science and Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
125
|
Chen Y, Aslanoglou S, Gervinskas G, Abdelmaksoud H, Voelcker NH, Elnathan R. Cellular Deformations Induced by Conical Silicon Nanowire Arrays Facilitate Gene Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1904819. [PMID: 31599099 DOI: 10.1002/smll.201904819] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/29/2019] [Indexed: 06/10/2023]
Abstract
Engineered cell-nanostructured interfaces generated by vertically aligned silicon nanowire (SiNW) arrays have become a promising platform for orchestrating cell behavior, function, and fate. However, the underlying mechanism in SiNW-mediated intracellular access and delivery is still poorly understood. This study demonstrates the development of a gene delivery platform based on conical SiNW arrays for mechanical cell transfection, assisted by centrifugal force, for both adherent and nonadherent cells in vitro. Cells form focal adhesions on SiNWs within 6 h, and maintain high viability and motility. Such a functional and dynamic cell-SiNW interface features conformational changes in the plasma membrane and in some cases the nucleus, promoting both direct penetration and endocytosis; this synergistically facilitates SiNW-mediated delivery of nucleic acids into immortalized cell lines, and into difficult-to-transfect primary immune T cells without pre-activation. Moreover, transfected cells retrieved from SiNWs retain the capacity to proliferate-crucial to future biomedical applications. The results indicate that SiNW-mediated intracellular delivery holds great promise for developing increasingly sophisticated investigative and therapeutic tools.
Collapse
Affiliation(s)
- Yaping Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, 3168, Australia
| | - Stella Aslanoglou
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, 3168, Australia
| | - Gediminas Gervinskas
- Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, 15 Innovation Walk, Clayton, VIC, 3800, Australia
| | - Hazem Abdelmaksoud
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, 5095, Australia
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, 3168, Australia
- INM-Leibniz Institute for New Materials, Campus D2 2, Saarbrücken, 66123, Germany
| | - Roey Elnathan
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| |
Collapse
|
126
|
Fazil MHUT, Chalasani MLS, Choong YK, Schmidtchen A, Verma NK, Saravanan R. A C-terminal peptide of TFPI-1 facilitates cytosolic delivery of nucleic acid cargo into mammalian cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1862:183093. [PMID: 31672541 DOI: 10.1016/j.bbamem.2019.183093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/16/2019] [Accepted: 10/10/2019] [Indexed: 12/12/2022]
Abstract
Efficient intracellular nucleic acid delivery into mammalian cells remains a long-standing challenge owing to poor cell permeability and uptake of naked nucleic acids across the cell membrane and limited cargo stability. Conventional delivery methods have several drawbacks, such as cytotoxicity, limited cell-type applicability, low efficiency, hindrances that limit the potential of oligonucleotide delivery in functional genomics, therapeutics and diverse research applications. Thus, new approaches that are robust, safe, effective and valid across multiple cell types are much needed. Here, we demonstrate that GGL27, a TFPI-1-derived novel cationic host defence peptide, facilitates the delivery of nucleic acid cargo into the cytosol of a range of mammalian cells. The GGL27 peptide is non-cytotoxic and is internalized in a broad range of mammalian cell-types, including transformed cell lines and primary cells. GGL27 spontaneously forms complexes with nucleic acids of variable sizes, protects them from nuclease degradation, and delivers cargo effectively. Together, our observations demonstrate the versatile cell-penetrating property of GGL27, providing an excellent template for developing a simple, non-toxic peptide-based cytosolic delivery tool for wide use in biomedical research.
Collapse
Affiliation(s)
| | | | - Yeu Khai Choong
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore
| | - Artur Schmidtchen
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden; Wound Healing Centre, Bispebjerg Hospital, Department of Biomedical Sciences, University of Copenhagen, Denmark
| | - Navin Kumar Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore; Skin Research Institute of Singapore, 11 Mandalay Road, Clinical Sciences Building, Singapore 308232, Singapore
| | - Rathi Saravanan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore.
| |
Collapse
|
127
|
Moore N, Chevillet JR, Healey LJ, McBrine C, Doty D, Santos J, Teece B, Truslow J, Mott V, Hsi P, Tandon V, Borenstein JT, Balestrini J, Kotz K. A Microfluidic Device to Enhance Viral Transduction Efficiency During Manufacture of Engineered Cellular Therapies. Sci Rep 2019; 9:15101. [PMID: 31641163 PMCID: PMC6806008 DOI: 10.1038/s41598-019-50981-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/23/2019] [Indexed: 02/08/2023] Open
Abstract
The development and approval of engineered cellular therapies are revolutionizing approaches to treatment of diseases. However, these life-saving therapies require extensive use of inefficient bioprocessing equipment and specialized reagents that can drive up the price of treatment. Integration of new genetic material into the target cells, such as viral transduction, is one of the most costly and labor-intensive steps in the production of cellular therapies. Approaches to reducing the costs associated with gene delivery have been developed using microfluidic devices to increase overall efficiency. However, these microfluidic approaches either require large quantities of virus or pre-concentration of cells with high-titer viral particles. Here, we describe the development of a microfluidic transduction device (MTD) that combines microfluidic spatial confinement with advective flow through a membrane to efficiently colocalize target cells and virus particles. We demonstrate that the MTD can improve the efficiency of lentiviral transduction for both T-cell and hematopoietic stem-cell (HSC) targets by greater than two fold relative to static controls. Furthermore, transduction saturation in the MTD is reached with only half the virus required to reach saturation under static conditions. Moreover, we show that MTD transduction does not adversely affect cell viability or expansion potential.
Collapse
Affiliation(s)
- Nathan Moore
- Cell and Tissue Engineering, 555 Technology Square, Draper, Cambridge, MA, 02139, USA.
| | - John R Chevillet
- Cell and Tissue Engineering, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Laura J Healey
- Cell and Tissue Engineering, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Connor McBrine
- Synthetic Biology, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Daniel Doty
- Cell and Tissue Engineering, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Jose Santos
- Biological Microsystems, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Bryan Teece
- Biological Microsystems, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - James Truslow
- Biological Microsystems, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Vienna Mott
- Biological Microsystems, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Peter Hsi
- Cell and Tissue Engineering, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Vishal Tandon
- Biological Microsystems, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | | | - Jenna Balestrini
- Biological Microsystems, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Kenneth Kotz
- Biological Microsystems, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| |
Collapse
|
128
|
A novel method to purify adenovirus based on increasing salt concentrations in buffer. Eur J Pharm Sci 2019; 141:105090. [PMID: 31626964 DOI: 10.1016/j.ejps.2019.105090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/02/2019] [Accepted: 09/24/2019] [Indexed: 02/05/2023]
Abstract
With the rapid development of gene therapy, gene-based medicine with adenovirus as vectors has become a new method for disease treatment. However, there are still enormous challenges in the large-scale production of adenoviruses for clinical use. Recent reports show that ion-exchange chromatography (IEC) is an effective tool for the isolation and purification of adenovirus. However, during the separation and purification, host cell protein and DNA, as well as serum from the culture medium, can non-specifically occupy numerous binding sites of the chromatography packings, thereby reducing the binding between the adenovirus and packing media. We here report a novel method for highly efficient purification of adenoviruses by increasing the salt concentrations of the samples to be ultrafiltrated by tangential flow filtration, the diafiltration buffer, and the samples for IEC purification. This method could significantly remove a large amount of serum proteins and host cell proteins, increase the amount of sample loaded on the IEC column, and improve the binding of the adenovirus samples to the packing media. A purity of > 95% could be obtained after one chromatography operation, and the number of purification steps and the amount of used packing media were reduced. The method is simple, economical, and efficient, and has excellent applications.
Collapse
|
129
|
Huo S, Gong N, Jiang Y, Chen F, Guo H, Gan Y, Wang Z, Herrmann A, Liang XJ. Gold-DNA nanosunflowers for efficient gene silencing with controllable transformation. SCIENCE ADVANCES 2019; 5:eaaw6264. [PMID: 31616782 PMCID: PMC6774715 DOI: 10.1126/sciadv.aaw6264] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 09/06/2019] [Indexed: 05/20/2023]
Abstract
The development of an efficient delivery system for enhanced and controlled gene interference-based therapeutics is still facing great challenges. Fortunately, the flourishing field of nanotechnology provides more effective strategies for nucleic acid delivery. Here, the triplex-forming oligonucleotide sequence and its complementary strand were used to mediate self-assembly of ultrasmall gold nanoparticles. The obtained sunflower-like nanostructures exhibited strong near-infrared (NIR) absorption and photothermal conversion ability. Upon NIR irradiation, the large-sized nanostructure could disassemble and generate ultrasmall nanoparticles modified with c-myc oncogene silencing sequence, which could directly target the cell nucleus. Moreover, the controlled gene silencing effect could be realized by synergistically controlling the preincubation time with the self-assembled nanostructure (in vitro and in vivo) and NIR irradiation time point. This study provides a new approach for constructing more efficient and tailorable nanocarriers for gene interference applications.
Collapse
Affiliation(s)
- Shuaidong Huo
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11 First North Road, Zhongguancun, Beijing 100190, P. R. China
- DWI–Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52056 Aachen, Germany
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- Corresponding author. (S.H.); (X.-J.L.)
| | - Ningqiang Gong
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11 First North Road, Zhongguancun, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Ying Jiang
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| | - Fei Chen
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Hongbo Guo
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11 First North Road, Zhongguancun, Beijing 100190, P. R. China
| | - Yaling Gan
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11 First North Road, Zhongguancun, Beijing 100190, P. R. China
| | - Zhisen Wang
- Shijiazhuang Zangnuo Bio Incorporated Corporation, No. 518 Cangsheng Road, Hi-tech District, Shijiazhuang, Hebei Province 050000, P. R. China
| | - Andreas Herrmann
- DWI–Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52056 Aachen, Germany
| | - Xing-Jie Liang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11 First North Road, Zhongguancun, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- Corresponding author. (S.H.); (X.-J.L.)
| |
Collapse
|
130
|
Gao YG, Lin X, Dang K, Jiang SF, Tian Y, Liu FL, Li DJ, Li Y, Miao ZP, Qian AR. Structure-activity relationship of novel low-generation dendrimers for gene delivery. Org Biomol Chem 2019; 16:7833-7842. [PMID: 30084471 DOI: 10.1039/c8ob01767k] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Structure-activity relationship (SAR) studies are very critical to design ideal gene vectors for gene delivery. However, It is difficult to obtain SAR information of low-generation dendrimers due to the lack of easy structural modification ways. Here, we synthesized a novel family of rigid aromatic backbone-based low-generation polyamidoamine (PAMAM) dendrimers. According to the number of primary amines, they were divided into two types: four-amine-containing PAMAM (DL1-DL5) and eight-amine-containing PAMAM (DL6-DL10). Due to the introduction of a rigid aromatic backbone, the low-generation PAMAM could be modified easier by different hydrophobic aliphatic chains. Several assays were used to study the interactions of the PAMAM dendrimers with plasmid DNA, and the results revealed that they not only had good DNA binding ability but also could efficiently condense DNA into spherical-shaped nanoparticles with suitable sizes and zeta potentials. The SAR studies indicated that the gene-transfection efficiency of the synthesized materials depended on not only the structure of their hydrophobic chains but also the number of primary amines. It was found that four-amine-containing PAMAM prepared from oleylamine (DL5) gave the best transfection efficiency, which was 3 times higher than that of lipofectamine 2000 in HEK293 cells. The cellular uptake mechanism mediated by DL5 was further investigated, and the results indicated that DL5/DNA complexes entered the cells mainly via caveolae and clathrin-mediated endocytosis. In addition, these low-generation PAMAMs modified with a single hydrophobic tail showed lower toxicity than lipofectamine 2000 in MC3T3-E1, MG63, HeLa, and HEK293 cells. These results reveal that such a type of low-generation polyamidoamines might be promising non-viral gene vectors, and also give us clues for the design of safe and high-efficiency gene vectors.
Collapse
Affiliation(s)
- Yong-Guang Gao
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi 710072, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Balakrishnan B, David E. Biopolymers augment viral vectors based gene delivery. J Biosci 2019; 44:84. [PMID: 31502562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The success of viral vectors mediated gene therapy is still hampered by immunogenicity and insufficient transgene expression. Alternatively, non-viral vectors mediated gene delivery has the advantage of low immunogenicity despite showing low transgene expression. By carefully considering the advantages of each approach, hybrid vectors are currently being developed by modifying the viral vectors using non-viral biopolymers. This review provides an overview of the hybrid vectors currently being developed.
Collapse
Affiliation(s)
- Balaji Balakrishnan
- Department of Haematology, Christian Medical College, Vellore 632004, Tamil Nadu, India
| | | |
Collapse
|
132
|
Zeng M, Alshehri F, Zhou D, Lara-Sáez I, Wang X, Li X, A S, Xu Q, Zhang J, Wang W. Efficient and Robust Highly Branched Poly(β-amino ester)/Minicircle COL7A1 Polymeric Nanoparticles for Gene Delivery to Recessive Dystrophic Epidermolysis Bullosa Keratinocytes. ACS APPLIED MATERIALS & INTERFACES 2019; 11:30661-30672. [PMID: 31390173 DOI: 10.1021/acsami.9b13135] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Recessive dystrophic epidermolysis bullosa (RDEB) is a severe congenital skin fragility disease caused by COL7A1 mutations that result in type VII collagen (C7) deficiency. Herein, we report a synergistic polyplex system that can efficiently restore C7 expression in RDEB keratinocytes. A highly branched multifunctional poly(β-amino ester) (HPAE), termed as HC32-122, was optimized systematically as the high-performance gene delivery vector for keratinocytes, achieving much higher transfection capability than polyethylenimine, SuperFect, and Lipofectamine 2000 without inducing obvious cytotoxicity. Concurrently, a 12 kb length minicircle DNA encoding ∼9 kb full-length COL7A1 (MCC7) devoid of bacterial sequence was biosynthesized as the therapeutic gene. Combining the highly potent polymer and the miniaturized gene structure, HC32-122/MCC7 polyplexes achieve 96.4% cellular uptake efficiency, 4019-fold COL7A1 mRNA enhancement, and robust recombinant C7 expression. Structure-property investigations reveal that HC32-122 can effectively condense MCC7 to form small, uniform, compact, and positively charged spherical nanoparticles with high DNA release flexibility. Moreover, formulation study shows that sucrose is conductive to lyophilized HC32-122/DNA polyplexes for maintaining the transfection capability. Direct frozen polyplexes can maintain full gene transfection capability after one-year storage. High efficiency, biocompatibility, facile manipulation, and long-term stability make the HC32-122/MCC7 system a promising bench-to-bed candidate for treating the debilitating RDEB.
Collapse
Affiliation(s)
- Ming Zeng
- Charles Institute of Dermatology , University College Dublin , Dublin D04 V1W8 , Ireland
- Department of Dermatology , The First Affiliated Hospital of Anhui Medical University , Hefei 230022 , China
| | - Fatma Alshehri
- Charles Institute of Dermatology , University College Dublin , Dublin D04 V1W8 , Ireland
- Princess Nourah bint Abdulrahman University , Riyadh 11671 , Saudi Arabia
| | - Dezhong Zhou
- Charles Institute of Dermatology , University College Dublin , Dublin D04 V1W8 , Ireland
- School of Chemical Engineering and Technology , Xi'an Jiaotong University , Xi'an , Shaanxi 710049 , China
| | - Irene Lara-Sáez
- Charles Institute of Dermatology , University College Dublin , Dublin D04 V1W8 , Ireland
| | - Xi Wang
- Charles Institute of Dermatology , University College Dublin , Dublin D04 V1W8 , Ireland
| | - Xiaolin Li
- Charles Institute of Dermatology , University College Dublin , Dublin D04 V1W8 , Ireland
| | - Sigen A
- Charles Institute of Dermatology , University College Dublin , Dublin D04 V1W8 , Ireland
| | - Qian Xu
- Charles Institute of Dermatology , University College Dublin , Dublin D04 V1W8 , Ireland
| | - Jing Zhang
- Charles Institute of Dermatology , University College Dublin , Dublin D04 V1W8 , Ireland
| | - Wenxin Wang
- Charles Institute of Dermatology , University College Dublin , Dublin D04 V1W8 , Ireland
| |
Collapse
|
133
|
Wu HY, Cao CY. The application of CRISPR-Cas9 genome editing tool in cancer immunotherapy. Brief Funct Genomics 2019; 18:129-132. [PMID: 29579146 DOI: 10.1093/bfgp/ely011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (CRISPR-Cas9) system was originally discovered in prokaryotes functioned as a part of the adaptive immune system. Because of its high efficiency and easy operability, CRISPR-Cas9 system has been developed to be a powerful and versatile gene editing tool shortly after its discovery. Given that multiple genetic alterations are the main factors that drive genesis and development of tumor, CRISPR-Cas9 system has been applied to correct cancer-causing gene mutations and deletions and to engineer immune cells, such as chimeric antigen receptor T (CAR T) cells, for cancer immunotherapeutic applications. Recently, CRISPR-Cas9-based CAR T-cell preparation has been an important breakthrough in antitumor therapy. Here, we summarize the mechanism, delivery and the application of CRISPR-Cas9 in gene editing, and discuss the challenges and future directions of CRISPR-Cas9 in cancer immunotherapy.
Collapse
Affiliation(s)
- Hong-Yan Wu
- Department of Immunology, Medical College, China Three Gorges University
| | - Chun-Yu Cao
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, Medical College, China Three Gorges University
| |
Collapse
|
134
|
|
135
|
Systemic delivery of Eg5 shRNA-expressing plasmids using PEGylated DC-Chol/DOPE cationic liposome: Long-term silencing and anticancer effects in vivo. Biochem Pharmacol 2019; 166:192-202. [DOI: 10.1016/j.bcp.2019.05.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 05/21/2019] [Indexed: 12/23/2022]
|
136
|
Puhl DL, D'Amato AR, Gilbert RJ. Challenges of gene delivery to the central nervous system and the growing use of biomaterial vectors. Brain Res Bull 2019; 150:216-230. [PMID: 31173859 PMCID: PMC8284997 DOI: 10.1016/j.brainresbull.2019.05.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 05/08/2019] [Accepted: 05/29/2019] [Indexed: 12/18/2022]
Abstract
Gene therapy is a promising form of treatment for those suffering from neurological disorders or central nervous system (CNS) injury, however, obstacles remain that limit its translational potential. The CNS is protected by the blood brain barrier, and this barrier blocks genes from traversing into the CNS if administered outside of the CNS. Viral and non-viral gene delivery vehicles, commonly referred to as vectors, are modified to enhance delivery efficiency to target locations in the CNS. Still, there are few gene therapy approaches approved by the FDA for CNS disease or injury treatment. The lack of viable clinical approaches is due, in part, to the unpredictable nature of many vector systems. In particular, safety concerns exist with the use of viral vectors for CNS gene delivery. To seek some alternatives to viral vectors, development of new non-viral, biomaterial vectors is occurring at a rapid rate. This review discusses the challenges of delivering various forms of genetic material to the CNS, the use and limitations of current viral vector delivery systems, and the use of non-viral, biomaterial vectors for CNS applications.
Collapse
Affiliation(s)
- Devan L Puhl
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, New York, 12180, United States; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th Street, Troy, New York, 12180, United States.
| | - Anthony R D'Amato
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, New York, 12180, United States; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th Street, Troy, New York, 12180, United States.
| | - Ryan J Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, New York, 12180, United States; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th Street, Troy, New York, 12180, United States.
| |
Collapse
|
137
|
Current Transport Systems and Clinical Applications for Small Interfering RNA (siRNA) Drugs. Mol Diagn Ther 2019; 22:551-569. [PMID: 29926308 DOI: 10.1007/s40291-018-0338-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Small interfering RNAs (siRNAs) are an attractive new agent with potential as a therapeutic tool because of its ability to inhibit specific genes for many conditions, including viral infections and cancers. However, despite this potential, many challenges remain, including off-target effects, difficulties with delivery, immune responses, and toxicity. Traditional genetic vectors do not guarantee that siRNAs will silence genes in vivo. Rational design strategies, such as chemical modification, viral vectors, and non-viral vectors, including cationic liposomes, polymers, nanocarriers, and bioconjugated siRNAs, provide important opportunities to overcome these challenges. We summarize the results of research into vector delivery of siRNAs as a therapeutic agent from their design to clinical trials in ophthalmic diseases, cancers, respiratory diseases, and liver virus infections. Finally, we discuss the current state of siRNA delivery methods and the need for greater understanding of the requirements.
Collapse
|
138
|
Killian T, Buntz A, Herlet T, Seul H, Mundigl O, Längst G, Brinkmann U. Antibody-targeted chromatin enables effective intracellular delivery and functionality of CRISPR/Cas9 expression plasmids. Nucleic Acids Res 2019; 47:e55. [PMID: 30809660 PMCID: PMC6547418 DOI: 10.1093/nar/gkz137] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/21/2019] [Accepted: 02/20/2019] [Indexed: 01/01/2023] Open
Abstract
We report a novel system for efficient and specific targeted delivery of large nucleic acids to and into cells. Plasmid DNA and core histones were assembled to chromatin by salt gradient dialysis and subsequently connected to bispecific antibody derivatives (bsAbs) via a nucleic acid binding peptide bridge. The resulting reconstituted vehicles termed 'plasmid-chromatin' deliver packaged nucleic acids to and into cells expressing antigens that are recognized by the bsAb, enabling intracellular functionality without detectable cytotoxicity. High efficiency of intracellular nucleic acid delivery is revealed by intracellular expression of plasmid encoded genes in most (∼90%) target cells to which the vehicles were applied under normal growth/medium conditions in nanomolar concentrations. Specific targeting, uptake and transgene expression depends on antibody-mediated cell surface binding: plasmid chromatin of identical composition but with non-targeting bsAbs or without bsAbs is ineffective. Examples that demonstrate applicability, specificity and efficacy of antibody-targeted plasmid chromatin include reporter gene constructs as well as plasmids that enable CRISPR/Cas9 mediated genome editing of target cells.
Collapse
Affiliation(s)
- Tobias Killian
- Roche Pharma Research and Early Development (pRED), Therapeutic Modalities - Large Molecule Research, Roche Innovation Center Munich, Nonnenwald 2, D-82377 Penzberg, Germany
| | - Annette Buntz
- Roche Pharma Research and Early Development (pRED), Therapeutic Modalities - Large Molecule Research, Roche Innovation Center Munich, Nonnenwald 2, D-82377 Penzberg, Germany
| | - Teresa Herlet
- Roche Pharma Research and Early Development (pRED), Therapeutic Modalities - Large Molecule Research, Roche Innovation Center Munich, Nonnenwald 2, D-82377 Penzberg, Germany
| | - Heike Seul
- Roche Pharma Research and Early Development (pRED), Therapeutic Modalities - Large Molecule Research, Roche Innovation Center Munich, Nonnenwald 2, D-82377 Penzberg, Germany
| | - Olaf Mundigl
- Roche Pharma Research and Early Development (pRED), Therapeutic Modalities - Large Molecule Research, Roche Innovation Center Munich, Nonnenwald 2, D-82377 Penzberg, Germany
| | - Gernot Längst
- Biochemistry III; Biochemistry Centre Regensburg (BCR), University of Regensburg, Regensburg, Germany
| | - Ulrich Brinkmann
- Roche Pharma Research and Early Development (pRED), Therapeutic Modalities - Large Molecule Research, Roche Innovation Center Munich, Nonnenwald 2, D-82377 Penzberg, Germany
| |
Collapse
|
139
|
Liu XQ, Xin HY, Lyu YN, Ma ZW, Peng XC, Xiang Y, Wang YY, Wu ZJ, Cheng JT, Ji JF, Zhong JX, Ren BX, Wang XW, Xin HW. Oncolytic herpes simplex virus tumor targeting and neutralization escape by engineering viral envelope glycoproteins. Drug Deliv 2019; 25:1950-1962. [PMID: 30799657 PMCID: PMC6282442 DOI: 10.1080/10717544.2018.1534895] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Oncolytic herpes simplex viruses (oHSVs) have been approved for clinical usage and become more and more popular for tumor virotherapy. However, there are still many issues for the oHSVs used in clinics and clinical trials. The main issues are the limited anti-tumor effects, intratumor injection, and some side effects. To overcome such challenges, here we review the genetic engineering of the envelope glycoproteins for oHSVs to target tumors specifically, and at the same time we summarize the many neutralization antibodies against the envelope glycoproteins and align the neutralization epitopes with functional domains of the respective glycoproteins for future identification of new functions of the glycoproteins and future engineering of the epitopes to escape from host neutralization.
Collapse
Affiliation(s)
- Xiao-Qin Liu
- a Faculty of Medicine, The Second School of Clinical Medicine , Yangtze University, Nanhuan , Jingzhou , Hubei , China.,b Laboratory of Oncology, Faculty of Medicine, Center for Molecular Medicine, School of Basic Medicine , Yangtze University , Jingzhou , Hubei , China.,c Faculty of Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medicine , Yangtze University , Jingzhou , Hubei , China.,d Department of Nursing and Medical Imaging Technology , Yangtze University , Jingzhou , Hubei , China
| | - Hong-Yi Xin
- e Star Array Pte Ltd , JTC Medtech Hub , Singapore , Singapore
| | - Yan-Ning Lyu
- f Institute for Infectious Diseases and Endemic Diseases Prevention and Control, Beijing Center for Diseases Prevention and Control , Beijing , China
| | - Zhao-Wu Ma
- a Faculty of Medicine, The Second School of Clinical Medicine , Yangtze University, Nanhuan , Jingzhou , Hubei , China.,b Laboratory of Oncology, Faculty of Medicine, Center for Molecular Medicine, School of Basic Medicine , Yangtze University , Jingzhou , Hubei , China.,c Faculty of Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medicine , Yangtze University , Jingzhou , Hubei , China
| | - Xiao-Chun Peng
- a Faculty of Medicine, The Second School of Clinical Medicine , Yangtze University, Nanhuan , Jingzhou , Hubei , China.,b Laboratory of Oncology, Faculty of Medicine, Center for Molecular Medicine, School of Basic Medicine , Yangtze University , Jingzhou , Hubei , China.,g Faculty of Medicine, Department of Pathophysiology, School of Basic Medicine , Yangtze University , Jingzhou , Hubei , China
| | - Ying Xiang
- a Faculty of Medicine, The Second School of Clinical Medicine , Yangtze University, Nanhuan , Jingzhou , Hubei , China.,b Laboratory of Oncology, Faculty of Medicine, Center for Molecular Medicine, School of Basic Medicine , Yangtze University , Jingzhou , Hubei , China.,c Faculty of Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medicine , Yangtze University , Jingzhou , Hubei , China
| | - Ying-Ying Wang
- a Faculty of Medicine, The Second School of Clinical Medicine , Yangtze University, Nanhuan , Jingzhou , Hubei , China.,b Laboratory of Oncology, Faculty of Medicine, Center for Molecular Medicine, School of Basic Medicine , Yangtze University , Jingzhou , Hubei , China.,c Faculty of Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medicine , Yangtze University , Jingzhou , Hubei , China
| | - Zi-Jun Wu
- a Faculty of Medicine, The Second School of Clinical Medicine , Yangtze University, Nanhuan , Jingzhou , Hubei , China.,b Laboratory of Oncology, Faculty of Medicine, Center for Molecular Medicine, School of Basic Medicine , Yangtze University , Jingzhou , Hubei , China.,c Faculty of Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medicine , Yangtze University , Jingzhou , Hubei , China.,d Department of Nursing and Medical Imaging Technology , Yangtze University , Jingzhou , Hubei , China
| | - Jun-Ting Cheng
- a Faculty of Medicine, The Second School of Clinical Medicine , Yangtze University, Nanhuan , Jingzhou , Hubei , China.,b Laboratory of Oncology, Faculty of Medicine, Center for Molecular Medicine, School of Basic Medicine , Yangtze University , Jingzhou , Hubei , China.,c Faculty of Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medicine , Yangtze University , Jingzhou , Hubei , China
| | - Jia-Fu Ji
- h Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery , Peking University Cancer Hospital and Institute , Haidian , Beijing , China
| | - Ji-Xin Zhong
- i Cardiovascular Research Institute , Case Western Reserve University , Cleveland , OH , USA
| | - Bo-Xu Ren
- a Faculty of Medicine, The Second School of Clinical Medicine , Yangtze University, Nanhuan , Jingzhou , Hubei , China.,d Department of Nursing and Medical Imaging Technology , Yangtze University , Jingzhou , Hubei , China
| | - Xian-Wang Wang
- a Faculty of Medicine, The Second School of Clinical Medicine , Yangtze University, Nanhuan , Jingzhou , Hubei , China.,b Laboratory of Oncology, Faculty of Medicine, Center for Molecular Medicine, School of Basic Medicine , Yangtze University , Jingzhou , Hubei , China.,j Faculty of Medicine, Department of Laboratory Medicine, School of Basic Medicine , Yangtze University , Jingzhou , Hubei , China
| | - Hong-Wu Xin
- a Faculty of Medicine, The Second School of Clinical Medicine , Yangtze University, Nanhuan , Jingzhou , Hubei , China.,b Laboratory of Oncology, Faculty of Medicine, Center for Molecular Medicine, School of Basic Medicine , Yangtze University , Jingzhou , Hubei , China.,c Faculty of Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medicine , Yangtze University , Jingzhou , Hubei , China
| |
Collapse
|
140
|
Yuan Y, Gu Z, Yao C, Luo D, Yang D. Nucleic Acid-Based Functional Nanomaterials as Advanced Cancer Therapeutics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1900172. [PMID: 30972963 DOI: 10.1002/smll.201900172] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/04/2019] [Indexed: 06/09/2023]
Abstract
Nucleic acid-based functional nanomaterials (NAFN) have been widely used as emerging drug delivery nanocarriers for cancer therapeutics. Considerable works have demonstrated that NAFN can effectively load and protect therapeutic agents, and particularly enable targeting delivery to the tumor site and stimuli-responsive release. These outstanding performances are due to NAFN's unique properties including inherent biological functions and sequence programmability as well as biocompatibility and biodegradability. In this Review, the recent progress on NAFN as advanced cancer therapeutics is highlighted. Three main cancer therapy approaches are categorized including chemo-, immuno-, and gene-therapy. Examples are presented to show how NAFN are rationally and exquisitely designed to address problems in cancer therapy. The challenges and future development of NAFN are also discussed toward future more practical biomedical applications.
Collapse
Affiliation(s)
- Ye Yuan
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, P. R. China
| | - Zi Gu
- School of Chemical Engineering and Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Chi Yao
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, P. R. China
| | - Dan Luo
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, P. R. China
| | - Dayong Yang
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, P. R. China
| |
Collapse
|
141
|
Lee S, Ahn HJ. Anti-EpCAM-conjugated adeno-associated virus serotype 2 for systemic delivery of EGFR shRNA: Its retargeting and antitumor effects on OVCAR3 ovarian cancer in vivo. Acta Biomater 2019; 91:258-269. [PMID: 31026519 DOI: 10.1016/j.actbio.2019.04.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 04/01/2019] [Accepted: 04/22/2019] [Indexed: 12/16/2022]
Abstract
Adeno-associated virus (AAV) is a promising vector for systemic delivery of siRNA because of its long-term expression ability without immunogenicity and pathogenicity. However, its broad host tropism and lack of tissue specificity have limited clinical applications such as cancer therapy. Therefore, redirecting the natural tropism of AAV vectors to unique cell surface antigens is an important requirement for in vivo RNAi-based cancer therapy. To use the overexpression property of epithelial cell adhesion molecule (EpCAM) in specific cancer types, we herein created anti-EpCAM antibody-conjugated AAV serotype 2 (AAV2) vectors through a streptavidin-biotin bridge. Upon intravenous injection, anti-EpCAM-conjugated AAV2 vectors showed prominent tumor-specific accumulation in EpCAM-positive tumor-bearing mice without undesirable sequestration in liver. In addition, when loaded with transgenes to express shRNA against epidermal growth factor receptor (EGFR), systemically injected anti-EpCAM-conjugated AAV2/shEGFR vectors induced significant downregulation of EGFR expression in tumors and eventually suppressed tumor growth even at the long dosing interval of two weeks. This in vivo antitumor effect represents the increased infection efficacy of tropism-modified AAV2 vectors and prolonged expression of EGFR shRNA in tumor tissues. Thus, this study suggests the great potential of anti-EpCAM-conjugated AAV2/shEGFR vectors as RNAi-based cancer therapeutics. STATEMENT OF SIGNIFICANCE: Adeno-associated virus (AAV) is a promising vector for systemic delivery of siRNA, but its broad host tropism has limited clinical applications. By using the overexpression property of epithelial cell adhesion molecule (EpCAM) on tumors, we demonstrate that anti-EpCAM-conjugated AAV2 vectors through a streptavidin-biotin bridge are redirected to EpCAM-positive tumors in vivo. In addition, when loaded with transgenes to express shRNA against epidermal growth factor receptor (EGFR), systemically injected anti-EpCAM-conjugated AAV2/shEGFR vectors significantly downregulate EGFR expression in tumors, eventually suppressing tumor growth for long periods. We herein suggest the potential of anti-EpCAM-AAV2/shEGFR vectors as an antitumor agent. Furthermore, redirection of AAV2 infection through EpCAM would provide a powerful means for systemic delivery of short hairpin RNA to tumor sites.
Collapse
|
142
|
Kant R, Bhatt G, Patel VK, Ganguli A, Singh D, Nayak M, Mishra K, Gupta A, Gangopadhyay K, Gangopadhyay S, Ramanathan G, Bhattacharya S. Synchronized Electromechanical Shock Wave-Induced Bacterial Transformation. ACS OMEGA 2019; 4:8512-8521. [PMID: 31459941 PMCID: PMC6648450 DOI: 10.1021/acsomega.9b00202] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 05/06/2019] [Indexed: 05/28/2023]
Abstract
We report a simple device that generates synchronized mechanical and electrical pressure waves for carrying out bacterial transformation. The mechanical pressure waves are produced by igniting a confined nanoenergetic composite material that provides ultrahigh pressure. Further, this device has an arrangement through which a synchronized electric field (of a time-varying nature) is initiated at a delay of ≈85 μs at the full width half-maxima point of the pressure pulse. The pressure waves so generated are incident to a thin aluminum-polydimethylsiloxane membrane that partitions the ignition chamber from the column of the mixture containing bacterial cells (Escherichia coli BL21) and 4 kb transforming DNA. A combination of mechanical and electrical pressure pulse created through the above arrangement ensures that the transforming DNA transports across the cell membrane into the cell, leading to a transformation event. This unique device has been successfully operated for efficient gene (∼4 kb) transfer into cells. The transformation efficacy of this device is found comparable to the other standard methods and protocols for carrying out the transformation.
Collapse
Affiliation(s)
- Rishi Kant
- Microsystems
Fabrication Laboratory, Department of Mechanical Engineering and Department of
Chemistry, Indian Institute of Technology
Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Geeta Bhatt
- Microsystems
Fabrication Laboratory, Department of Mechanical Engineering and Department of
Chemistry, Indian Institute of Technology
Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Vinay Kumar Patel
- Microsystems
Fabrication Laboratory, Department of Mechanical Engineering and Department of
Chemistry, Indian Institute of Technology
Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Anurup Ganguli
- Microsystems
Fabrication Laboratory, Department of Mechanical Engineering and Department of
Chemistry, Indian Institute of Technology
Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Deepak Singh
- Microsystems
Fabrication Laboratory, Department of Mechanical Engineering and Department of
Chemistry, Indian Institute of Technology
Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Monalisha Nayak
- Microsystems
Fabrication Laboratory, Department of Mechanical Engineering and Department of
Chemistry, Indian Institute of Technology
Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Keerti Mishra
- Microsystems
Fabrication Laboratory, Department of Mechanical Engineering and Department of
Chemistry, Indian Institute of Technology
Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Ankur Gupta
- Microsystems
Fabrication Laboratory, Department of Mechanical Engineering and Department of
Chemistry, Indian Institute of Technology
Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Keshab Gangopadhyay
- Department
of Electrical and Computer Engineering, University of Missouri Columbia, Columbia, Missouri 65211, United States
| | - Shubhra Gangopadhyay
- Department
of Electrical and Computer Engineering, University of Missouri Columbia, Columbia, Missouri 65211, United States
| | - Gurunath Ramanathan
- Microsystems
Fabrication Laboratory, Department of Mechanical Engineering and Department of
Chemistry, Indian Institute of Technology
Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Shantanu Bhattacharya
- Microsystems
Fabrication Laboratory, Department of Mechanical Engineering and Department of
Chemistry, Indian Institute of Technology
Kanpur, Kanpur 208016, Uttar Pradesh, India
| |
Collapse
|
143
|
Therapeutic application of the CRISPR system: current issues and new prospects. Hum Genet 2019; 138:563-590. [PMID: 31115652 DOI: 10.1007/s00439-019-02028-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 05/13/2019] [Indexed: 12/23/2022]
|
144
|
Kizer ME, Deng Y, Kang G, Mikael PE, Wang X, Chung AJ. Hydroporator: a hydrodynamic cell membrane perforator for high-throughput vector-free nanomaterial intracellular delivery and DNA origami biostability evaluation. LAB ON A CHIP 2019; 19:1747-1754. [PMID: 30964485 DOI: 10.1039/c9lc00041k] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The successful intracellular delivery of exogenous macromolecules is crucial for a variety of applications ranging from basic biology to the clinic. However, traditional intracellular delivery methods such as those relying on viral/non-viral nanocarriers or physical membrane disruptions suffer from low throughput, toxicity, and inconsistent delivery performance and are time-consuming and/or labor-intensive. In this study, we developed a single-step hydrodynamic cell deformation-induced intracellular delivery platform named "hydroporator" without the aid of vectors or a complicated/costly external apparatus. By utilizing only fluid inertia, the platform focuses, guides, and stretches cells robustly without clogging. This rapid hydrodynamic cell deformation leads to both convective and diffusive delivery of external (macro)molecules into the cell through transient plasma membrane discontinuities. Using this hydroporation approach, highly efficient (∼90%), high-throughput (>1 600 000 cells per min), and rapid delivery (∼1 min) of different (macro)molecules into a wide range of cell types was achieved while maintaining high cell viability. Taking advantage of the ability of this platform to rapidly deliver large molecules, we also systematically investigated the temporal biostability of vanilla DNA origami nanostructures in living cells for the first time. Experiments using two DNA origami (tube- and donut-shaped) nanostructures revealed that these nanostructures can maintain their structural integrity in living cells for approximately 1 h after delivery, providing new opportunities for the rapid characterization of intracellular DNA biostability.
Collapse
Affiliation(s)
- Megan E Kizer
- Department of Chemistry and Chemical Biology, Centre for Biotechnology and Interdisciplinary Studies (CBIS), Rensselaer Polytechnic Institute (RPI), Troy, NY 12180, USA.
| | | | | | | | | | | |
Collapse
|
145
|
Bisht DS, Bhatia V, Bhattacharya R. Improving plant-resistance to insect-pests and pathogens: The new opportunities through targeted genome editing. Semin Cell Dev Biol 2019; 96:65-76. [PMID: 31039395 DOI: 10.1016/j.semcdb.2019.04.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/09/2019] [Accepted: 04/12/2019] [Indexed: 12/26/2022]
Abstract
The advantages of high input agriculture are fading away due to degenerating soil health and adverse effects of climate change. Safeguarding crop yields in the changing environment and dynamics of pest and pathogens, has posed new challenges to global agriculture. Thus, integration of new technologies in crop improvement has been imperative for achieving the breeding objectives in faster ways. Recently, enormous potential of genome editing through engineered nucleases has been demonstrated in plants. Continuous refinements of the genome editing tools have increased depth and breadth of their applications. So far, genome editing has been demonstrated in more than fifty plant species. These include model species like Arabidopsis, as well as important crops like rice, wheat, maize etc. Particularly, CRISPR/Cas9 based two component genome editing system has been facile with wider applicability. Potential of genome editing has unfurled enormous possibilities for engineering diverse agronomic traits including durable resistance against insect-pests and pathogens. Novel propositions of developing insect and pathogen resistant crops by genome editing include altering the effector-target interaction, knocking out of host-susceptibility genes, engineering synthetic immune receptor eliciting broad spectrum resistance, uncoupling of antagonistic action of defense hormones etc. Alternatively, modification of insect genomes has been used either to create gene drive or to counteract resistance to various insecticides. The distinct advantage of genome editing system is that it can knock out specific target region in the genome without leaving the unwanted vector backbone. In this article, we have reviewed the novel opportunities offered by the genome editing technologies for developing insect and pathogen resistant crop-types, their future prospects and anticipated challenges.
Collapse
Affiliation(s)
- Deepak Singh Bisht
- ICAR-National Institute for Plant Biotechnology, Indian Agricultural Research Institute Campus, New Delhi, India
| | - Varnika Bhatia
- Deen Dayal Upadhyaya College, University of Delhi, Delhi, India
| | - Ramcharan Bhattacharya
- ICAR-National Institute for Plant Biotechnology, Indian Agricultural Research Institute Campus, New Delhi, India.
| |
Collapse
|
146
|
Chen MY, Butler SS, Chen W, Suh J. Physical, chemical, and synthetic virology: Reprogramming viruses as controllable nanodevices. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1545. [PMID: 30411529 PMCID: PMC6461522 DOI: 10.1002/wnan.1545] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 08/03/2018] [Accepted: 10/04/2018] [Indexed: 01/24/2023]
Abstract
The fields of physical, chemical, and synthetic virology work in partnership to reprogram viruses as controllable nanodevices. Physical virology provides the fundamental biophysical understanding of how virus capsids assemble, disassemble, display metastability, and assume various configurations. Chemical virology considers the virus capsid as a chemically addressable structure, providing chemical pathways to modify the capsid exterior, interior, and subunit interfaces. Synthetic virology takes an engineering approach, modifying the virus capsid through rational, combinatorial, and bioinformatics-driven design strategies. Advances in these three subfields of virology aim to develop virus-based materials and tools that can be applied to solve critical problems in biomedicine and biotechnology, including applications in gene therapy and drug delivery, diagnostics, and immunotherapy. Examples discussed include mammalian viruses, such as adeno-associated virus (AAV), plant viruses, such as cowpea mosaic virus (CPMV), and bacterial viruses, such as Qβ bacteriophage. Importantly, research efforts in physical, chemical, and synthetic virology have further unraveled the design principles foundational to the form and function of viruses. This article is categorized under: Diagnostic Tools > Diagnostic Nanodevices Biology-Inspired Nanomaterials > Protein and Virus-Based Structures.
Collapse
Affiliation(s)
| | - Susan S Butler
- Department of Bioengineering, Rice University, Houston, Texas
| | - Weitong Chen
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, Texas
| | - Junghae Suh
- Department of Bioengineering, Rice University, Houston, Texas
- Systems, Synthetic, and Physical Biology Program, Rice University, Houston, Texas
| |
Collapse
|
147
|
Li P, Wang J, Chen G, Zhang X, Lin D, Zhou Y, Yu Y, Liu W, Zhang D. Oncolytic activity of canine distemper virus in canine mammary tubular adenocarcinoma cells. Vet Comp Oncol 2019; 17:174-183. [PMID: 30756476 DOI: 10.1111/vco.12466] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/31/2019] [Accepted: 02/06/2019] [Indexed: 12/24/2022]
Abstract
Canine distemper virus (CDV), bearing a close resemblance to measles virus, represents a promising candidate for oncolytic therapy; however, its application and underlying oncolytic mechanisms in canine mammary carcinoma cells remain to be explored. Here, we found that an attenuated canine distemper vaccine strain, CDV-L, efficiently infected and inhibited the growth of canine mammary tubular adenocarcinoma CIPp cells but not MDCK cells in vitro. Transcriptomic analysis of CDV-L-infected CIPp cells revealed substantially differentially expressed genes in apoptotic and NF-κB signalling pathways. Subsequent validations confirmed that CDV-L-induced apoptosis of CIPp cells through the caspase-8 and caspase-3 pathway. Identification of phosphorylated-IκBα, phosphorylated-p65 and the nuclear translocation of p65 confirmed the activation of the NF-κB signalling pathway. Inhibition of the NF-κB pathway abrogated CDV-L-induced cleaved-caspase-3 and cleaved-PARP. In a CIPp subcutaneous xenograft mouse model, intratumoural injections of CDV-L significantly restricted tumour growth without apparent pathology, and virus remained localized within the tumour. Taken altogether, these findings indicate that CDV-L exerts an antitumour effect in CIPp cells, and that apoptosis and the NF-κB pathway play essential roles in this process.
Collapse
Affiliation(s)
- Peiran Li
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, P.R. China
| | - Jigui Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, P.R. China
| | - Gaoxiang Chen
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, P.R. China
| | - Xiaomei Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, P.R. China
| | - Degui Lin
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, P.R. China
| | - Yun Zhou
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, P.R. China
| | - Yongle Yu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, P.R. China
| | - Weiquan Liu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, P.R. China
| | - Di Zhang
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, P.R. China
| |
Collapse
|
148
|
Pottash AE, Kuffner C, Noonan-Shueh M, Jay SM. Protein-based vehicles for biomimetic RNAi delivery. J Biol Eng 2019; 13:19. [PMID: 30891095 PMCID: PMC6390323 DOI: 10.1186/s13036-018-0130-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 12/09/2018] [Indexed: 12/30/2022] Open
Abstract
Broad translational success of RNA interference (RNAi) technology depends on the development of effective delivery approaches. To that end, researchers have developed a variety of strategies, including chemical modification of RNA, viral and non-viral transfection approaches, and incorporation with delivery vehicles such as polymer- and lipid-based nanoparticles, engineered and native proteins, extracellular vesicles (EVs), and others. Among these, EVs and protein-based vehicles stand out as biomimetically-inspired approaches, as both proteins (e.g. Apolipoprotein A-1, Argonaute 2, and Arc) and EVs mediate intercellular RNA transfer physiologically. Proteins specifically offer significant therapeutic potential due to their biophysical and biochemical properties as well as their ability to facilitate and tolerate manipulation; these characteristics have made proteins highly successful translational therapeutic molecules in the last two decades. This review covers engineered protein vehicles for RNAi delivery along with what is currently known about naturally-occurring extracellular RNA carriers towards uncovering design rules that will inform future engineering of protein-based vehicles.
Collapse
Affiliation(s)
- Alex Eli Pottash
- 1Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742 USA
| | - Christopher Kuffner
- 1Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742 USA
| | - Madeleine Noonan-Shueh
- 1Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742 USA
| | - Steven M Jay
- 1Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742 USA.,2Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA.,3Program in Molecular and Cellular Biology, University of Maryland, College Park, MD 20742 USA
| |
Collapse
|
149
|
Joshi PR, Cervera L, Ahmed I, Kondratov O, Zolotukhin S, Schrag J, Chahal PS, Kamen AA. Achieving High-Yield Production of Functional AAV5 Gene Delivery Vectors via Fedbatch in an Insect Cell-One Baculovirus System. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 13:279-289. [PMID: 30886878 PMCID: PMC6404649 DOI: 10.1016/j.omtm.2019.02.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 02/07/2019] [Indexed: 11/19/2022]
Abstract
Despite numerous advancements in production protocols, manufacturing AAV to meet exceptionally high demand (1016–1017 viral genomes [VGs]) in late clinical stages and for eventual systemic delivery poses significant challenges. Here, we report an efficient, simple, scalable, robust AAV5 production process utilizing the most recent modification of the OneBac platform. An increase in volumetric yield of genomic particles by ∼6-fold and functional particles by ∼20-fold was achieved by operating a high-cell-density process in shake flasks and bioreactors that involves an Sf9-based rep/cap stable cell line grown at a density of about 10 million cells/mL infected with a single baculovirus. The overall volumetric yields of genomic (VG) and bioactive particles (enhanced transducing units [ETUs]) in representative fedbatch bioreactor runs ranged from 2.5 to 3.5 × 1014 VG/L and from 1 to 2 × 1011 ETU/L. Analytical ultracentrifugation analyses of affinity-purified AAV vector samples from side-by-side batch and fedbatch production runs showed vector preparations with a full and empty particle distribution of 20%–30% genomic and 70%–80% empty particles. Moreover, the stoichiometric analysis of capsid proteins from fedbatch production in shake flask and bioreactor run samples demonstrated the incorporation of higher VP1 subunits, resulting in better functionality.
Collapse
Affiliation(s)
- Pranav R.H. Joshi
- Viral Vectors and Vaccine Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada
| | - Laura Cervera
- Viral Vectors and Vaccine Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada
| | - Ibrahim Ahmed
- Viral Vectors and Vaccine Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada
| | - Oleksandr Kondratov
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Sergei Zolotukhin
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Joseph Schrag
- Human Health Therapeutics Portfolio, National Research Council of Canada, Montreal, QC H4P 2R2, Canada
| | - Parminder S. Chahal
- Human Health Therapeutics Portfolio, National Research Council of Canada, Montreal, QC H4P 2R2, Canada
| | - Amine A. Kamen
- Viral Vectors and Vaccine Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada
- Corresponding author: Amine Kamen, Viral Vectors and Vaccine Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada.
| |
Collapse
|
150
|
Karimian A, Azizian K, Parsian H, Rafieian S, Shafiei‐Irannejad V, Kheyrollah M, Yousefi M, Majidinia M, Yousefi B. CRISPR/Cas9 technology as a potent molecular tool for gene therapy. J Cell Physiol 2019; 234:12267-12277. [PMID: 30697727 DOI: 10.1002/jcp.27972] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 11/19/2018] [Indexed: 12/30/2022]
Affiliation(s)
- Ansar Karimian
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences Babol Iran
- Cancer & Immunology Research Center, Kurdistan University of Medical Sciences Sanandaj Iran
- Student Research Committee, Babol University of Medical Sciences Babol Iran
| | - Khalil Azizian
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Science Tabriz Iran
| | - Hadi Parsian
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences Babol Iran
| | - Sona Rafieian
- Department of Oral and Maxillofacial Pathology School of Dentistry, Zanjan University of Medical Sciences Zanjan Iran
| | | | - Maryam Kheyrollah
- Department of Molecular Medicine National Institue of Genetic Engeneering and Biotechnology Tehran Iran
| | - Mehdi Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
| | - Maryam Majidinia
- Tumor Research Center, Urmia University of Medical Sciences Urmia Iran
| | - Bahman Yousefi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences Tehran Iran
- Department of Biochemistry and Clinical Laboratories Faculty of Medicine, Tabriz University of Medical Science Tabriz Iran
| |
Collapse
|