1
|
Soh WWM, Finol E, Chan SJW, Zhu JY, Liau SSJK, Bier A, Ooi EE, Bazan GC. Tailoring Lipid Nanoparticle with Ex Situ Incorporated Conjugated Oligoelectrolyte for Enhanced mRNA Delivery Efficiency. Adv Healthc Mater 2025; 14:e2405048. [PMID: 40103511 DOI: 10.1002/adhm.202405048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/20/2025] [Indexed: 03/20/2025]
Abstract
Developing new lipid nanoparticle (LNP) formulations typically involves reconstruction from separate elements followed by rigorous purification steps, contributing to drawn-out drug discovery processes. Membrane-intercalating conjugated oligoelectrolytes (COEs) are water-soluble molecules featuring a conjugated backbone and peripheral ionic groups, specifically designed to spontaneously integrate into lipid bilayers. Herein, an ex situ strategy to "dope" the representative COE-S6 into pre-formed messenger RNA-LNPs (mRNA-LNPs) is presented, exploiting its spontaneous membrane intercalation property through a straightforward add-and-mix procedure. Incorporating 0.2% COE-S6 into mRNA-LNPs relative to lipid content reduced particle size from 84.5 ± 1 to 67.9 ± 0.8 nm, elevated cellular uptake, and improved endosomal escape. These traits culminate in an increase in in cellula transfection from 24.2 ± 1.6% to 98.7 ± 0.6%. When injected intravenously into healthy BALB/c mice, the optimized COE-S6-doped mRNA-LNPs boost in vivo luciferase expression by 1.75-fold. Additionally, COE-S6-doped mRNA-LNPs exhibit fluorogenic properties, enabling intracellular mechanistic studies via confocal microscopy. This simple method enhances the properties of mRNA-LNPs with minimal COE quantities, offering a novel strategy to improve existing LNP formulations and provide optical reporting capabilities, essential for expediting drug discovery and delivery.
Collapse
Affiliation(s)
- Wilson Wee Mia Soh
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
| | - Esteban Finol
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, National University of Singapore, Singapore, 169857, Singapore
| | - Samuel J W Chan
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
| | - Ji-Yu Zhu
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
| | | | - Ava Bier
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
| | - Eng Eong Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, National University of Singapore, Singapore, 169857, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, 169857, Singapore
| | - Guillermo C Bazan
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
| |
Collapse
|
2
|
Setegne M, Cabral AT, Tiwari A, Shen F, Thiam HR, Dassama LMK. Engineering Cell-Specific Protein Delivery Vehicles for Erythroid Lineage Cells. ACS BIO & MED CHEM AU 2025; 5:268-282. [PMID: 40255284 PMCID: PMC12006860 DOI: 10.1021/acsbiomedchemau.4c00098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 02/11/2025] [Accepted: 02/11/2025] [Indexed: 04/22/2025]
Abstract
Biologics such as proteins, peptides, and oligonucleotides are powerful ligands to modulate challenging drug targets that lack readily accessible and "ligandable" pockets. However, the limited membrane permeance of biologics severely restricts their intracellular applications. Moreover, different cell types may exhibit varying levels of impermeability, and some delivery vehicles might be more sensitive to this variance. Erythroid lineage cells are especially challenging to deliver cargo to because of their unique cytoskeleton and the absence of endocytosis in mature erythrocytes. We recently employed a cell permeant miniature protein to deliver bioPROTACs to human umbilical cord blood derived erythroid progenitor cells (HUDEP-2) and primary hematopoietic stem (CD34+) cells (Shen et al., ACS Cent. Sci.2022, 8, 1695-1703). While successful, the low efficiency of delivery and lack of cell-type specificity limit use of bioPROTACs in vivo. In this work, we thoroughly evaluated the performance of various recently reported cell penetrating peptides (CPPs), CPP additives, bacterial toxins, and contractile injection systems for their ability to deliver cargo to erythroid precursor cells. We also explored how targeting receptors enriched on the erythroid cell surface might improve the efficiencies and specificities of these delivery vehicles. Our results reveal that certain vehicles exhibit improved efficiencies when directed to cell surface receptors while others do not benefit from this targeting strategy. Together, these findings advance our understanding of protein delivery to challenging cell types and illustrate some of the intricacies of cell-surface receptor targeting.
Collapse
Affiliation(s)
- Mekedlawit
T. Setegne
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
- Sarafan
ChEM-H Institute, Stanford University, Stanford, California 94305, United States
| | - Aidan T. Cabral
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
| | - Anushri Tiwari
- Department
of Biology, Stanford University, Stanford, California 94305, United States
| | - Fangfang Shen
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Hawa Racine Thiam
- Sarafan
ChEM-H Institute, Stanford University, Stanford, California 94305, United States
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
- Department
of Microbiology & Immunology, Stanford
School of Medicine, Stanford, California 94305, United States
| | - Laura M. K. Dassama
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
- Sarafan
ChEM-H Institute, Stanford University, Stanford, California 94305, United States
- Department
of Microbiology & Immunology, Stanford
School of Medicine, Stanford, California 94305, United States
| |
Collapse
|
3
|
Duong T, Firmo M, Li CT, Gu B, Wang P. Three-dimensional linkage analysis with digital PCR for genome integrity and identity of recombinant adeno-associated virus. Sci Rep 2025; 15:2154. [PMID: 39820513 PMCID: PMC11739598 DOI: 10.1038/s41598-024-77378-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 10/22/2024] [Indexed: 01/19/2025] Open
Abstract
Recombinant adeno-associated virus (rAAV) has emerged as the vector of choice for in vivo gene delivery, with numerous clinical trials underway for the treatment of various human diseases. Utilizing rAAV in gene therapy requires a highly precise quantification method to determine the viral genome titer and further establish the optimal therapeutic dosage for a rAAV product. The conventional single-channel droplet digital PCR (1D ddPCR) method offers only partial information regarding the viral vector genome titer, lacking insights into its integrity. In our pursuit of further advancing rAAV analysis, we have developed a novel 3D ddPCR assay with advanced 3D linkage analysis. We have designed the three amplicon sites targeting both ends of the viral genome, as well as the center of key therapeutic gene of interest (GOI). This study aims to offer a more comprehensive and insightful assessment of rAAV products which includes not only quantity of viral genome titer but also the quality, distinguishing between partial ones and intact full-length viral genomes with the right GOI. Importantly, due to the random partitioning property of a digital PCR system, the 3D linkage analysis of rAAV viral genome requires a proper mathematical model to identify the true linked DNA molecules (full-length/intact DNA) from the population of false/unlinked DNA molecules (fragmented/partial DNA). We therefore have developed an AAV 3D linkage analysis workflow to characterize genomic integrity and intact titer for rAAV gene therapy products. In this study, we focus on evaluating our 3D linkage mathematical model by performing DNA mixing experiments and a case study using multiple rAAV samples. Particularly, we rigorously tested our algorithms by conducting experiments involving the mixing of seven DNA fragments to represent various AAV viral genome populations, including 3 single partials, 3 double partials, and 1 full-length genomes. Across all 37 tested scenarios, we validated the accuracy of our workflow's output for the percentages of 3D linkage by comparing to the known percentages of input DNA. Consequently, our comprehensive AAV analytical package not only offers insights into viral genome titer but also provides valuable information on its integrity and identity. This cost-effective approach, akin to the setup of traditional 1D or 2D dPCR, holds the potential to advance the application of rAAV in cell and gene therapy for the treatment of human diseases.
Collapse
Affiliation(s)
- Tam Duong
- Research & Development, Lonza Houston, Inc., 14905 Kirby Dr, Houston, TX, 77047, USA.
| | - Michele Firmo
- Global Biologics Technical Development, Lonza Basel, Inc., Muenchensteinerstrasse 38, 4052, Basel, Switzerland
| | - Chien-Ting Li
- Research & Development, Lonza Houston, Inc., 14905 Kirby Dr, Houston, TX, 77047, USA
| | - Bingnan Gu
- Research & Development, Lonza Houston, Inc., 14905 Kirby Dr, Houston, TX, 77047, USA.
| | - Peng Wang
- Research & Development, Lonza Houston, Inc., 14905 Kirby Dr, Houston, TX, 77047, USA.
| |
Collapse
|
4
|
Alissa M, Aldurayhim M, Abdulaziz O, Alsalmi O, Awad A, Algopishi UB, Alharbi S, Safhi AY, Khan KH, Uffar C. From molecules to heart regeneration: Understanding the complex and profound role of non-coding RNAs in stimulating cardiomyocyte proliferation for cardiac repair. Curr Probl Cardiol 2024; 49:102857. [PMID: 39306148 DOI: 10.1016/j.cpcardiol.2024.102857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024]
Abstract
Recent studies of noncoding genomes have shown important implications for regulating gene expression and genetic programs during development and their association with health, including cardiovascular disease. There are nearly 2,500 microRNAs (miRNAs), 12,000 long-chain non-coding RNAs (lncRNA), and nearly 4,000 circular RNAs (circles). Even though they do not code for proteins, they make up nearly 99% of the human genome. Non-coding RNA families (ncRNAs) have recently been discovered and established as novel and necessary controllers of cardiovascular risk factors and cellular processes and, therefore, have the potential to improve the diagnosis and prediction of cardiovascular disease. The increase in the prevalence of cardiovascular disease can be explained by the shortcomings of existing therapies, which focus only on the non-coding RNAs that protein codes for. On the other hand, recent studies point to the possibility of using ncRNAs in the early detection and intervention of CVD. These findings suggest that developing diagnostic tools and therapies based on miRNAs, lncRNAs, and circRNAs will potentially enhance the clinical management of patients with cardiovascular disease. Cardiovascular diseases include CH, HF, RHD, ACS, MI, AS, MF, ARR, and PAH, of which CH is the most common cardiovascular disease, followed by HF and RHD. This paper aims to elucidate the biological and clinical significance of miRNAs, increase, and circles, as well as their expression profiles and the possibility of regulating non-coding transcripts in cardiovascular diseases to improve the application of ncRNAs in diagnosis and treatment.
Collapse
Affiliation(s)
- Mohammed Alissa
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
| | - Mohammed Aldurayhim
- Department of Medical Laboratory, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Osama Abdulaziz
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21974, Saudi Arabia
| | - Ohud Alsalmi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21974, Saudi Arabia
| | - Alsamghan Awad
- King Khalid University, College of Medicine, Family Medicine department, Saudi Arabia
| | | | - Sarah Alharbi
- Department of Medical Laboratory, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Awaji Y Safhi
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Khadijah Hassan Khan
- Department of Laboratory, King Faisal Medical Complex, Ministry of Health, Taif 26514, Saudi Arabia
| | - Christin Uffar
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| |
Collapse
|
5
|
Wang R, Wang Z, Tong L, Wang R, Yao S, Chen D, Hu H. Microfluidic Mechanoporation: Current Progress and Applications in Stem Cells. BIOSENSORS 2024; 14:256. [PMID: 38785730 PMCID: PMC11117831 DOI: 10.3390/bios14050256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/08/2024] [Accepted: 05/12/2024] [Indexed: 05/25/2024]
Abstract
Intracellular delivery, the process of transporting substances into cells, is crucial for various applications, such as drug delivery, gene therapy, cell imaging, and regenerative medicine. Among the different approaches of intracellular delivery, mechanoporation stands out by utilizing mechanical forces to create temporary pores on cell membranes, enabling the entry of substances into cells. This method is promising due to its minimal contamination and is especially vital for stem cells intended for clinical therapy. In this review, we explore various mechanoporation technologies, including microinjection, micro-nano needle arrays, cell squeezing through physical confinement, and cell squeezing using hydrodynamic forces. Additionally, we highlight recent research efforts utilizing mechanoporation for stem cell studies. Furthermore, we discuss the integration of mechanoporation techniques into microfluidic platforms for high-throughput intracellular delivery with enhanced transfection efficiency. This advancement holds potential in addressing the challenge of low transfection efficiency, benefiting both basic research and clinical applications of stem cells. Ultimately, the combination of microfluidics and mechanoporation presents new opportunities for creating comprehensive systems for stem cell processing.
Collapse
Affiliation(s)
- Rubing Wang
- Zhejiang University-University of Illinois Urbana-Champaign Institute (ZJU-UIUC Institute), International Campus, Haining 314400, China;
| | - Ziqi Wang
- Center for Regeneration and Cell Therapy of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China; (Z.W.); (L.T.)
| | - Lingling Tong
- Center for Regeneration and Cell Therapy of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China; (Z.W.); (L.T.)
| | - Ruoming Wang
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), International Campus, Zhejiang University, Haining 314400, China; (R.W.); (S.Y.)
| | - Shuo Yao
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), International Campus, Zhejiang University, Haining 314400, China; (R.W.); (S.Y.)
| | - Di Chen
- Center for Regeneration and Cell Therapy of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China; (Z.W.); (L.T.)
- Center for Reproductive Medicine, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310003, China
- National Key Laboratory of Biobased Transportation Fuel Technology, Haining 314400, China
| | - Huan Hu
- Zhejiang University-University of Illinois Urbana-Champaign Institute (ZJU-UIUC Institute), International Campus, Haining 314400, China;
| |
Collapse
|
6
|
Gao X, Dong D, Zhang C, Deng Y, Ding J, Niu S, Tan S, Sun L. Chitosan-Functionalized Poly(β-Amino Ester) Hybrid System for Gene Delivery in Vaginal Mucosal Epithelial Cells. Pharmaceutics 2024; 16:154. [PMID: 38276521 PMCID: PMC10818660 DOI: 10.3390/pharmaceutics16010154] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Gene therapy displays great promise in the treatment of cervical cancer. The occurrence of cervical cancer is highly related to persistent human papilloma virus (HPV) infection. The HPV oncogene can be cleaved via gene editing technology to eliminate carcinogenic elements. However, the successful application of the gene therapy method depends on effective gene delivery into the vagina. To improve mucosal penetration and adhesion ability, quaternized chitosan was introduced into the poly(β-amino ester) (PBAE) gene-delivery system in the form of quaternized chitosan-g-PBAE (QCP). At a mass ratio of PBAE:QCP of 2:1, the polymers exhibited the highest green fluorescent protein (GFP) transfection efficiency in HEK293T and ME180 cells, which was 1.1 and 5.4 times higher than that of PEI 25 kD. At this mass ratio, PBAE-QCP effectively compressed the GFP into spherical polyplex nanoparticles (PQ-GFP NPs) with a diameter of 255.5 nm. In vivo results indicated that owing to the mucopenetration and adhesion capability of quaternized CS, the GFP transfection efficiency of the PBAE-QCP hybrid system was considerably higher than those of PBAE and PEI 25 kD in the vaginal epithelial cells of Sprague-Dawley rats. Furthermore, the new system demonstrated low toxicity and good safety, laying an effective foundation for its further application in gene therapy.
Collapse
Affiliation(s)
- Xueqin Gao
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Dirong Dong
- Department of Obstetrics and Gynecology, Zhongnan Hospital, Wuhan University, Wuhan 430062, China;
| | - Chong Zhang
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (C.Z.); (Y.D.); (J.D.); (S.N.)
- Health Bureau of Luannan Country, Tangshan 063599, China
| | - Yuxing Deng
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (C.Z.); (Y.D.); (J.D.); (S.N.)
| | - Jiahui Ding
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (C.Z.); (Y.D.); (J.D.); (S.N.)
| | - Shiqi Niu
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (C.Z.); (Y.D.); (J.D.); (S.N.)
| | - Songwei Tan
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (C.Z.); (Y.D.); (J.D.); (S.N.)
| | - Lili Sun
- Department of Obstetrics and Gynecology, Zhongnan Hospital, Wuhan University, Wuhan 430062, China;
| |
Collapse
|
7
|
Deng L, Han X, Wang Z, Nie X, Bian J. The Landscape of Noncoding RNA in Pulmonary Hypertension. Biomolecules 2022; 12:biom12060796. [PMID: 35740920 PMCID: PMC9220981 DOI: 10.3390/biom12060796] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 02/08/2023] Open
Abstract
The transcriptome of pulmonary hypertension (PH) is complex and highly genetically heterogeneous, with noncoding RNA transcripts playing crucial roles. The majority of RNAs in the noncoding transcriptome are long noncoding RNAs (lncRNAs) with less circular RNAs (circRNAs), which are two characteristics gaining increasing attention in the forefront of RNA research field. These noncoding transcripts (especially lncRNAs and circRNAs) exert important regulatory functions in PH and emerge as potential disease biomarkers and therapeutic targets. Recent technological advancements have established great momentum for discovery and functional characterization of ncRNAs, which include broad transcriptome sequencing such as bulk RNA-sequence, single-cell and spatial transcriptomics, and RNA-protein/RNA interactions. In this review, we summarize the current research on the classification, biogenesis, and the biological functions and molecular mechanisms of these noncoding RNAs (ncRNAs) involved in the pulmonary vascular remodeling in PH. Furthermore, we highlight the utility and challenges of using these ncRNAs as biomarkers and therapeutics in PH.
Collapse
Affiliation(s)
- Lin Deng
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (L.D.); (Z.W.)
| | - Xiaofeng Han
- Department of Diagnostic and Interventional Radiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China;
| | - Ziping Wang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (L.D.); (Z.W.)
| | - Xiaowei Nie
- Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People’s Hospital (The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518055, China
- Correspondence: (X.N.); (J.B.)
| | - Jinsong Bian
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (L.D.); (Z.W.)
- Correspondence: (X.N.); (J.B.)
| |
Collapse
|
8
|
Kwon DH, Gim GM, Eom KH, Lee JH, Jang G. Application of transposon systems in the transgenesis of bovine somatic and germ cells. BMC Vet Res 2022; 18:156. [PMID: 35477562 PMCID: PMC9044889 DOI: 10.1186/s12917-022-03252-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 04/14/2022] [Indexed: 12/12/2022] Open
Abstract
Background Several DNA transposons including PiggyBac (PB), Sleeping Beauty (SB), and Tol2 have been applied as effective means for of transgenesis in many species. Cattle are not typically experimental animals, and relatively little verification has been presented on this species. Thus, the goal here was to determine the applicability of three transposon systems in somatic and embryo cells in cattle, while also investigating which of the three systems is appropriate for each cell type. Green fluorescent protein (GFP)-expressing transposon systems were used for electroporation and microinjection in the somatic cells and embryo stage, respectively. After transfection, the GFP-positive cells or blastocysts were observed through fluorescence, while the transfection efficiency was calculated by FACS. Results In bovine somatic cells, the PB (63.97 ± 11.56) showed the highest efficiency of the three systems (SB: 50.74 ± 13.02 and Tol2: 16.55 ± 5.96). Conversely, Tol2 (75.00%) and SB (70.00%) presented a higher tendency in the embryonic cells compared to PB (42.86%). Conclusions These results demonstrate that these three transposon systems can be used in bovine somatic cells and embryos as gene engineering experimental methods. Moreover, they demonstrate which type of transposon system to apply depending on the cell type. Supplementary Information The online version contains supplementary material available at 10.1186/s12917-022-03252-1.
Collapse
Affiliation(s)
- Dong-Hyeok Kwon
- Laboratory of Theriogenology, College of Veterinary Medicine, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea, 08826.,BK21 PLUS program, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Gyeong-Min Gim
- Laboratory of Theriogenology, College of Veterinary Medicine, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea, 08826.,BK21 PLUS program, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kyeong-Hyeon Eom
- Laboratory of Theriogenology, College of Veterinary Medicine, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea, 08826.,BK21 PLUS program, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | | | - Goo Jang
- Laboratory of Theriogenology, College of Veterinary Medicine, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea, 08826. .,BK21 PLUS program, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea. .,LARTBio Inc, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Gangadaran P, Oh JM, Rajendran RL, Ahn BC. In Vivo Bioluminescent Imaging of Bone Marrow-Derived Mesenchymal Stem Cells in Mice. Methods Mol Biol 2022; 2525:281-287. [PMID: 35836076 DOI: 10.1007/978-1-0716-2473-9_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Mesenchymal stem cells (MSCs) are multipotent adult stem cells present in multiple tissues, such as bone marrow, adipose tissue, umbilical cord, and amniotic fluid. MSCs can differentiate into multilineage cells under defined conditions in vitro and in vivo. MSCs have been shown to have therapeutic effects on various types of diseases. Noninvasive in vivo monitoring of MSCs is considered one of the important techniques for developing cell therapy. In this protocol, we introduce strategized MSCs derived from bone marrow (BM-MSCs) of knock-in mouse model expressing mCherry-Renilla luciferase (mCherry-RLuc) for noninvasive bioluminescence imaging (BLI) of injected BM-MSCs in vivo.
Collapse
Affiliation(s)
- Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Byeong-Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea.
| |
Collapse
|
10
|
Mancinelli S, Turcato A, Kisslinger A, Bongiovanni A, Zazzu V, Lanati A, Liguori GL. Design of transfections: Implementation of design of experiments for cell transfection fine tuning. Biotechnol Bioeng 2021; 118:4488-4502. [PMID: 34406655 PMCID: PMC9291525 DOI: 10.1002/bit.27918] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/02/2021] [Accepted: 08/12/2021] [Indexed: 11/25/2022]
Abstract
Transfection is the process by which nucleic acids are introduced into eukaryotic cells. This is fundamental in basic research for studying gene function and modulation of gene expression as well as for many bioprocesses in the manufacturing of clinical‐grade recombinant biologics from cells. Transfection efficiency is a critical parameter to increase biologics' productivity; the right protocol has to be identified to ensure high transfection efficiency and therefore high product yield. Design of experiments (DoE) is a mathematical method that has become a key tool in bioprocess development. Based on the DoE method, we developed an operational flow that we called “Design of Transfections” (DoT) for specific transfection modeling and identification of the optimal transfection conditions. As a proof of principle, we applied the DoT workflow to optimize a cell transfection chemical protocol for neural progenitors, using polyethyleneimine (PEI). We simultaneously varied key influencing factors, namely concentration and type of PEI, DNA concentration, and cell density. The transfection efficiency was measured by fluorescence imaging followed by automatic counting of the green fluorescent transfected cells. Taking advantage of the DoT workflow, we developed a new simple, efficient, and economically advantageous PEI transfection protocol through which we were able to obtain a transfection efficiency of 34%.
Collapse
Affiliation(s)
- Sara Mancinelli
- Institute of Genetics and Biophysics (IGB), National Research Council (CNR), Naples, Italy
| | | | - Annamaria Kisslinger
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council (CNR), Naples, Italy
| | - Antonella Bongiovanni
- Institute for Research and Biomedical Innovation (IRIB), National Research Council (CNR), Palermo, Italy
| | - Valeria Zazzu
- Institute of Genetics and Biophysics (IGB), National Research Council (CNR), Naples, Italy
| | | | - Giovanna Lucia Liguori
- Institute of Genetics and Biophysics (IGB), National Research Council (CNR), Naples, Italy
| |
Collapse
|
11
|
Wichert N, Witt M, Blume C, Scheper T. Clinical applicability of optogenetic gene regulation. Biotechnol Bioeng 2021; 118:4168-4185. [PMID: 34287844 DOI: 10.1002/bit.27895] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 05/27/2021] [Accepted: 07/13/2021] [Indexed: 11/10/2022]
Abstract
The field of optogenetics is rapidly growing in relevance and number of developed tools. Among other things, the optogenetic repertoire includes light-responsive ion channels and methods for gene regulation. This review will be confined to the optogenetic control of gene expression in mammalian cells as suitable models for clinical applications. Here optogenetic gene regulation might offer an excellent method for spatially and timely regulated gene and protein expression in cell therapeutic approaches. Well-known systems for gene regulation, such as the LOV-, CRY2/CIB-, PhyB/PIF-systems, as well as other, in mammalian cells not yet fully established systems, will be described. Advantages and disadvantages with regard to clinical applications are outlined in detail. Among the many unanswered questions concerning the application of optogenetics, we discuss items such as the use of exogenous chromophores and their effects on the biology of the cells and methods for a gentle, but effective gene transfection method for optogenetic tools for in vivo applications.
Collapse
Affiliation(s)
- Nina Wichert
- Insitute of Technical Chemistry, Leibniz University of Hannover, Hannover, Germany
| | - Martin Witt
- Insitute of Technical Chemistry, Leibniz University of Hannover, Hannover, Germany
| | - Cornelia Blume
- Insitute of Technical Chemistry, Leibniz University of Hannover, Hannover, Germany
| | - Thomas Scheper
- Insitute of Technical Chemistry, Leibniz University of Hannover, Hannover, Germany
| |
Collapse
|
12
|
Kumar S, Li A, Thadhani NN, Prausnitz MR. Optimization of intracellular macromolecule delivery by nanoparticle-mediated photoporation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 37:102431. [PMID: 34175453 DOI: 10.1016/j.nano.2021.102431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 05/11/2021] [Accepted: 05/26/2021] [Indexed: 11/19/2022]
Abstract
Nanoparticle-mediated photoporation is a novel delivery platform for intracellular molecule delivery. We studied the dependence of macromolecular delivery on molecular weight and sought to enhance delivery efficiency. DU145 prostate cancer cells were exposed to pulsed laser beam in the presence of carbon-black nanoparticles. Intracellular uptake of molecules decreased with increasing molecular weight. Attributing this dependence to molecular diffusivity, we hypothesized that macromolecular delivery efficiency could be enhanced by increasing either laser fluence or laser exposure duration at low fluence. We observed increased percentages of macromolecule uptake by cells in both cases. However, trade-off between cell uptake and viability loss was most favorable at low laser fluence (25-29 mJ/cm2) and longer exposure durations (4-5 min). We conclude that long exposure at low laser fluence optimizes intracellular macromolecule delivery by nanoparticle-mediated photoporation, which may be explained by longer time for macromolecules to diffuse into cells, during and between laser pulses.
Collapse
Affiliation(s)
- Simple Kumar
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrew Li
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Naresh N Thadhani
- School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Mark R Prausnitz
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
13
|
Kumar S, Lazau E, Kim C, N Thadhani N, R Prausnitz M. Serum Protects Cells and Increases Intracellular Delivery of Molecules by Nanoparticle-Mediated Photoporation. Int J Nanomedicine 2021; 16:3707-3724. [PMID: 34103912 PMCID: PMC8180297 DOI: 10.2147/ijn.s307027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/12/2021] [Indexed: 12/03/2022] Open
Abstract
Introduction Intracellular delivery of molecules is central to applications in biotechnology, medicine, and basic research. Nanoparticle-mediated photoporation using carbon black nanoparticles exposed to pulsed, near-infrared laser irradiation offers a physical route to create transient cell membrane pores, enabling intracellular delivery. However, nanoparticle-mediated photoporation, like other physical intracellular delivery technologies, necessitates a trade-off between achieving efficient uptake of exogenous molecules and maintaining high cell viability. Methods In this study, we sought to shift this balance by adding serum to cells during nanoparticle-mediated photoporation as a viability protectant. DU-145 prostate cancer cells and human dermal fibroblasts were exposed to laser irradiation in the presence of carbon black (CB) nanoparticles and other formulation additives, including fetal bovine serum (FBS) and polymers. Results Our studies showed that FBS can protect cells from viability loss, even at high-fluence laser irradiation conditions that lead to high levels of intracellular delivery in two different mammalian cell types. Further studies revealed that full FBS was not needed: viability protection was achieved with denatured FBS, with just the high molecular weight fraction of FBS (>30 kDa), or even with individual proteins like albumin or hemoglobin. Finally, we found that viability protection was also obtained using certain neutral water-soluble polymers, including Pluronic F127, polyvinylpyrrolidone, poly(2-ethyl-2-oxazoline), and polyethylene glycol, which were more effective at increased concentration, molecular weight, or hydrophobicity. Conclusion Altogether, these findings suggest an interaction between amphiphilic domains of polymers with the cell membrane to help cells maintain viability, possibly by facilitating transmembrane pore closure. In this way, serum components or synthetic polymers can be used to increase intracellular delivery by nanoparticle-mediated photoporation while maintaining high cell viability.
Collapse
Affiliation(s)
- Simple Kumar
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Eunice Lazau
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30318, USA
| | - Carter Kim
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30318, USA
| | - Naresh N Thadhani
- School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Mark R Prausnitz
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| |
Collapse
|
14
|
Chong ZX, Yeap SK, Ho WY. Transfection types, methods and strategies: a technical review. PeerJ 2021; 9:e11165. [PMID: 33976969 PMCID: PMC8067914 DOI: 10.7717/peerj.11165] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 03/05/2021] [Indexed: 12/17/2022] Open
Abstract
Transfection is a modern and powerful method used to insert foreign nucleic acids into eukaryotic cells. The ability to modify host cells' genetic content enables the broad application of this process in studying normal cellular processes, disease molecular mechanism and gene therapeutic effect. In this review, we summarized and compared the findings from various reported literature on the characteristics, strengths, and limitations of various transfection methods, type of transfected nucleic acids, transfection controls and approaches to assess transfection efficiency. With the vast choices of approaches available, we hope that this review will help researchers, especially those new to the field, in their decision making over the transfection protocol or strategy appropriate for their experimental aims.
Collapse
Affiliation(s)
- Zhi Xiong Chong
- School of Pharmacy, University of Nottingham Malaysia, Semenyih, Selangor, Malaysia
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, Sepang, Selangor, Malaysia
| | - Wan Yong Ho
- School of Pharmacy, University of Nottingham Malaysia, Semenyih, Selangor, Malaysia
| |
Collapse
|
15
|
Morshedi Rad D, Alsadat Rad M, Razavi Bazaz S, Kashaninejad N, Jin D, Ebrahimi Warkiani M. A Comprehensive Review on Intracellular Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2005363. [PMID: 33594744 DOI: 10.1002/adma.202005363] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/22/2020] [Indexed: 05/22/2023]
Abstract
Intracellular delivery is considered an indispensable process for various studies, ranging from medical applications (cell-based therapy) to fundamental (genome-editing) and industrial (biomanufacture) approaches. Conventional macroscale delivery systems critically suffer from such issues as low cell viability, cytotoxicity, and inconsistent material delivery, which have opened up an interest in the development of more efficient intracellular delivery systems. In line with the advances in microfluidics and nanotechnology, intracellular delivery based on micro- and nanoengineered platforms has progressed rapidly and held great promises owing to their unique features. These approaches have been advanced to introduce a smorgasbord of diverse cargoes into various cell types with the maximum efficiency and the highest precision. This review differentiates macro-, micro-, and nanoengineered approaches for intracellular delivery. The macroengineered delivery platforms are first summarized and then each method is categorized based on whether it employs a carrier- or membrane-disruption-mediated mechanism to load cargoes inside the cells. Second, particular emphasis is placed on the micro- and nanoengineered advances in the delivery of biomolecules inside the cells. Furthermore, the applications and challenges of the established and emerging delivery approaches are summarized. The topic is concluded by evaluating the future perspective of intracellular delivery toward the micro- and nanoengineered approaches.
Collapse
Affiliation(s)
- Dorsa Morshedi Rad
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Maryam Alsadat Rad
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Sajad Razavi Bazaz
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Navid Kashaninejad
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Dayong Jin
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute of Molecular Medicine, Sechenov University, Moscow, 119991, Russia
| |
Collapse
|
16
|
Fazal S, Bisserier M, Hadri L. Molecular and Genetic Profiling for Precision Medicines in Pulmonary Arterial Hypertension. Cells 2021; 10:638. [PMID: 33805595 PMCID: PMC7999465 DOI: 10.3390/cells10030638] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/11/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare and chronic lung disease characterized by progressive occlusion of the small pulmonary arteries, which is associated with structural and functional alteration of the smooth muscle cells and endothelial cells within the pulmonary vasculature. Excessive vascular remodeling is, in part, responsible for high pulmonary vascular resistance and the mean pulmonary arterial pressure, increasing the transpulmonary gradient and the right ventricular "pressure overload", which may result in right ventricular (RV) dysfunction and failure. Current technological advances in multi-omics approaches, high-throughput sequencing, and computational methods have provided valuable tools in molecular profiling and led to the identification of numerous genetic variants in PAH patients. In this review, we summarized the pathogenesis, classification, and current treatments of the PAH disease. Additionally, we outlined the latest next-generation sequencing technologies and the consequences of common genetic variants underlying PAH susceptibility and disease progression. Finally, we discuss the importance of molecular genetic testing for precision medicine in PAH and the future of genomic medicines, including gene-editing technologies and gene therapies, as emerging alternative approaches to overcome genetic disorders in PAH.
Collapse
Affiliation(s)
| | | | - Lahouaria Hadri
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA; (S.F.); (M.B.)
| |
Collapse
|
17
|
Hasan MM, Ragnarsson L, Cardoso FC, Lewis RJ. Transfection methods for high-throughput cellular assays of voltage-gated calcium and sodium channels involved in pain. PLoS One 2021; 16:e0243645. [PMID: 33667217 PMCID: PMC7935312 DOI: 10.1371/journal.pone.0243645] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 11/25/2020] [Indexed: 11/24/2022] Open
Abstract
Chemical transfection is broadly used to transiently transfect mammalian cells, although often associated with cellular stress and membrane instability, which imposes challenges for most cellular assays, including high-throughput (HT) assays. In the current study, we compared the effectiveness of calcium phosphate, FuGENE and Lipofectamine 3000 to transiently express two key voltage-gated ion channels critical in pain pathways, CaV2.2 and NaV1.7. The expression and function of these channels were validated using two HT platforms, the Fluorescence Imaging Plate Reader FLIPRTetra and the automated patch clamp QPatch 16X. We found that all transfection methods tested demonstrated similar effectiveness when applied to FLIPRTetra assays. Lipofectamine 3000-mediated transfection produced the largest peak currents for automated patch clamp QPatch assays. However, the FuGENE-mediated transfection was the most effective for QPatch assays as indicated by the superior number of cells displaying GΩ seal formation in whole-cell patch clamp configuration, medium to large peak currents, and higher rates of accomplished assays for both CaV2.2 and NaV1.7 channels. Our findings can facilitate the development of HT automated patch clamp assays for the discovery and characterization of novel analgesics and modulators of pain pathways, as well as assisting studies examining the pharmacology of mutated channels.
Collapse
Affiliation(s)
- Md. Mahadhi Hasan
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Lotten Ragnarsson
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Fernanda C. Cardoso
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
- * E-mail: (FCC); (RJL)
| | - Richard J. Lewis
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
- * E-mail: (FCC); (RJL)
| |
Collapse
|
18
|
Gisbert Roca F, André FM, Más Estellés J, Monleón Pradas M, Mir LM, Martínez-Ramos C. BDNF-Gene Transfected Schwann Cell-Assisted Axonal Extension and Sprouting on New PLA-PPy Microfiber Substrates. Macromol Biosci 2021; 21:e2000391. [PMID: 33645917 DOI: 10.1002/mabi.202000391] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/11/2021] [Indexed: 01/09/2023]
Abstract
The work here reported analyzes the effect of increased efficiency of brain-derived neurotrophic factor (BDNF) production by electroporated Schwann cells (SCs) on the axonal extension in a coculture system on a biomaterial platform that can be of interest for the treatment of injuries of the nervous system, both central and peripheral. Rat SCs are electrotransfected with a plasmid coding for the BDNF protein in order to achieve an increased expression and release of this protein into the culture medium of the cells, performing the best balance between the level of transfection and the number of living cells. Gene-transfected SCs show an about 100-fold increase in the release of BDNF into the culture medium, compared to nonelectroporated SCs. Cocultivation of electroporated SCs with rat dorsal root ganglia (DRG) is performed on highly aligned substrates of polylactic acid (PLA) microfibers coated with the electroconductive polymer polypyrrol (PPy). The coculture of DRG with electrotransfected SCs increase both the axonal extension and the axonal sprouting from DRG neurons compared to the coculture of DRG with nonelectroporated SCs. Therefore, the use of PLA-PPy highly aligned microfiber substrates preseeded with electrotransfected SCs with an increased BDNF secretion is capable of both guiding and accelerating axonal growth.
Collapse
Affiliation(s)
- Fernando Gisbert Roca
- Centro de Biomateriales e Ingeniería Tisular, Universitat Politècnica de València, Camino de Vera s/n, Valencia, 46022, Spain
| | - Franck M André
- Metabolic and systemic aspects of oncogenesis (METSY), CNRS, Université Paris-Saclay, Institut Gustave Roussy, Villejuif, 94805, France
| | - Jorge Más Estellés
- Centro de Biomateriales e Ingeniería Tisular, Universitat Politècnica de València, Camino de Vera s/n, Valencia, 46022, Spain
| | - Manuel Monleón Pradas
- Centro de Biomateriales e Ingeniería Tisular, Universitat Politècnica de València, Camino de Vera s/n, Valencia, 46022, Spain.,CIBER-BBN, Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina, Madrid, 28029, Spain
| | - Lluis M Mir
- Metabolic and systemic aspects of oncogenesis (METSY), CNRS, Université Paris-Saclay, Institut Gustave Roussy, Villejuif, 94805, France
| | - Cristina Martínez-Ramos
- Centro de Biomateriales e Ingeniería Tisular, Universitat Politècnica de València, Camino de Vera s/n, Valencia, 46022, Spain.,Unitat predepartamental de Medicina, Universitat Jaume I, Avda/Sos Baynat, S/N, Castellón de la Plana, 12071, Spain
| |
Collapse
|
19
|
Dong X, Dong X, Gao F, Liu N, Liang T, Zhang F, Fu X, Pu L, Chen J. Non-coding RNAs in cardiomyocyte proliferation and cardiac regeneration: Dissecting their therapeutic values. J Cell Mol Med 2021; 25:2315-2332. [PMID: 33492768 PMCID: PMC7933974 DOI: 10.1111/jcmm.16300] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/05/2021] [Accepted: 01/09/2021] [Indexed: 12/23/2022] Open
Abstract
Cardiovascular diseases are associated with high incidence and mortality, contribute to disability and place a heavy economic burden on countries worldwide. Stimulating endogenous cardiomyocyte proliferation and regeneration has been considering as a key to repair the injured heart caused by ischaemia. Emerging evidence has proved that non‐coding RNAs participate in cardiac proliferation and regeneration. In this review, we focus on the observation and mechanism that microRNAs (or miRNAs), long non‐coding RNAs (or lncRNAs) and circular RNA (or circRNAs) regulate cardiomyocyte proliferation and regeneration to repair a damaged heart. Furthermore, we highlight the potential therapeutic role of some non‐coding RNAs used in stimulating CMs proliferation. Finally, perspective on the development of non‐coding RNAs therapy in cardiac regeneration is presented.
Collapse
Affiliation(s)
- Xiaoxuan Dong
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiuyun Dong
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan, China
| | - Feng Gao
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Ning Liu
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Tian Liang
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Zhang
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuyang Fu
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Linbin Pu
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinghai Chen
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
20
|
Liu D, Zhu M, Zhang Y, Diao Y. Crossing the blood-brain barrier with AAV vectors. Metab Brain Dis 2021; 36:45-52. [PMID: 33201426 DOI: 10.1007/s11011-020-00630-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/12/2020] [Indexed: 12/26/2022]
Abstract
Central nervous system (CNS) diseases are some of the most difficult to treat because the blood-brain barrier (BBB) almost entirely limits the passage of many therapeutic drugs into the CNS. Gene therapy based on the adeno-associated virus (AAV) vector has the potential to overcome this problem. For example, an AAV serotype AAV9 has been widely studied for its ability to cross the BBB to transduce astrocytes, but its efficiency is limited. The emergence of AAV directed evolution technology provides a solution, and the variants derived from AAV9 directed evolution have been shown to have significantly higher crossing efficiency than AAV9. However, the mechanisms by which AAV crosses the BBB are still unclear. In this review, we focus on recent advances in crossing the blood-brain barrier with AAV vectors. We first review the AAV serotypes that can be applied to treating CNS diseases. Recent progress in possible AAV crossing the BBB and transduction mechanisms are then summarized. Finally, the methods to improve the AAV transduction efficiency are discussed.
Collapse
Affiliation(s)
- Dan Liu
- School of Biomedical Sciences, College of Chemical Engineering, Huaqiao University, Xiamen, Fujian, China.
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China.
| | - Mingyang Zhu
- School of Biomedical Sciences, College of Chemical Engineering, Huaqiao University, Xiamen, Fujian, China
| | - Yuqian Zhang
- School of Biomedical Sciences, College of Chemical Engineering, Huaqiao University, Xiamen, Fujian, China
| | - Yong Diao
- School of Biomedical Sciences, College of Chemical Engineering, Huaqiao University, Xiamen, Fujian, China
| |
Collapse
|
21
|
Zhang SK, Gong L, Zhang X, Yun ZM, Li SB, Gao HW, Dai CJ, Yuan JJ, Chen JM, Gong F, Tan YX, Ji SP. Antimicrobial peptide AR-23 derivatives with high endosomal disrupting ability enhance poly(l-lysine)-mediated gene transfer. J Gene Med 2020; 22:e3259. [PMID: 32776410 PMCID: PMC7685122 DOI: 10.1002/jgm.3259] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/06/2020] [Accepted: 08/06/2020] [Indexed: 01/24/2023] Open
Abstract
Background pH‐sensitive peptides are a relatively new strategy for conquering the poor endosomal release of cationic polymer‐mediated transfection. Modification of antimicrobial peptides by exchanging positively‐charged residues with negatively‐charged glutamic acid residues (Glu) greatly improves its lytic activity at the endosomal pH, which could improve cationic polymer‐mediated transfection. Methods In the present study, we investigated the effect of the number of Glu substituted for positively‐charged residues on the endosomal escape activity of AR‐23 and the ability of mutated AR‐23 with respect to enhancing cationic polymer‐mediated transfection. Three analogs were synthesized by replacing the positively‐charged residues in the AR‐23 sequence with Glu one‐by‐one. Results The pH‐sensitive lysis ability of the peptides, the effect of peptides on the physicochemical characteristics, the intracellular trafficking, the transfection efficiency and the cytotoxicity of the polyplexes were determined. Increased lytic activity of peptides was observed with the increased number of Glu replacement in the AR‐23 sequence at acidic pH. The number of Glu substituted for positively‐charged residues of AR‐23 dramatically affects its lysis ability at neutral pH. Triple‐Glu substitution in the AR‐23 sequence greatly improved poly(l‐lysine)‐mediated gene transfection efficiency at the same time as maintaining low cytotoxicity. Conclusions The results indicate that replacement of positively‐charged residues with sufficient Glu residues may be considered as a method for designing pH‐sensitive peptides, which could be applied as potential enhancers for improving cationic polymer‐mediated transfection.
Collapse
Affiliation(s)
- Shi-Kun Zhang
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Lin Gong
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China.,PLA navy No. 971 Hospital, Qingdao, Shandong, China
| | - Xue Zhang
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Zhi-Min Yun
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Su-Bo Li
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Hong-Wei Gao
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Cong-Jie Dai
- College of Oceanology and Food Sciences, Quanzhou Normal University, Quanzhou, Fujian, China
| | - Jian-Jun Yuan
- College of Oceanology and Food Sciences, Quanzhou Normal University, Quanzhou, Fujian, China
| | - Jing-Ming Chen
- Quanzhou Preschool Education College, Quanzhou, Fujian, China
| | - Feng Gong
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Ying-Xia Tan
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Shou-Ping Ji
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China.,College of Oceanology and Food Sciences, Quanzhou Normal University, Quanzhou, Fujian, China
| |
Collapse
|
22
|
Bisserier M, Pradhan N, Hadri L. Current and emerging therapeutic approaches to pulmonary hypertension. Rev Cardiovasc Med 2020; 21:163-179. [PMID: 32706206 PMCID: PMC7389678 DOI: 10.31083/j.rcm.2020.02.597] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 05/25/2020] [Indexed: 12/15/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and fatal lung disease of multifactorial etiology. Most of the available drugs and FDA-approved therapies for treating pulmonary hypertension attempt to overcome the imbalance between vasoactive and vasodilator mediators, and restore the endothelial cell function. Traditional medications for treating PAH include the prostacyclin analogs and receptor agonists, phosphodiesterase 5 inhibitors, endothelin-receptor antagonists, and cGMP activators. While the current FDA-approved drugs showed improvements in quality of life and hemodynamic parameters, they have shown only very limited beneficial effects on survival and disease progression. None of them offers a cure against PAH, and the median survival rate remains less than three years from diagnosis. Extensive research efforts have led to the emergence of innovative therapeutic approaches in the area of PAH. In this review, we provide an overview of the current FDA-approved therapies in PAH and discuss the associated clinical trials and reported-side effects. As recent studies have led to the emergence of innovative therapeutic approaches in the area of PAH, we also focus on the latest promising therapies in preclinical studies such as stem cell-based therapies, gene transfer, and epigenetic therapies.
Collapse
Affiliation(s)
- Malik Bisserier
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Natasha Pradhan
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lahouaria Hadri
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
23
|
Blackstock DJ, Goh A, Shetty S, Fabozzi G, Yang R, Ivleva VB, Schwartz R, Horwitz J. Comprehensive Flow Cytometry Analysis of PEI-Based Transfections for Virus-Like Particle Production. RESEARCH 2020; 2020:1387402. [PMID: 32259105 PMCID: PMC7094759 DOI: 10.34133/2020/1387402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 01/28/2020] [Indexed: 11/06/2022]
Abstract
The generation of stable clones for biomolecule production is a common but lengthy and labor-intensive process. For complex molecules, such as viruses or virus-like particles (VLPs), the timeline becomes even more cumbersome. Thus, in the early stages of development, transient production methods serve as a reasonable alternative to stable clone construction. In this work, an investigation of a polyethylenimine- (PEI-) based transfection method for the transient production of Chikungunya (Chik) VLPs, a vaccine candidate molecule, was undertaken. This effort focuses on tracking cell population responses during transfection, understanding how process changes affect these responses, and monitoring patterns in cell performance over the culture duration. Plasmid labeling and VLP staining were employed to comprehensively track cells via flow cytometry and to draw correlations between plasmid DNA (pDNA) uptake and the resulting VLP expression. The method detected high transfection efficiency (≥97%) in all samples tested and demonstrated the capability to track kinetics of plasmid-cell binding. With varied transfection cell concentrations, the pDNA binding kinetics are altered and saturation binding is observed in the lowest cell concentration sample tested in less than 3 hours of incubation. Interestingly, in all samples, the flow cytometry analysis of relative pDNA amount versus VLP expression staining showed that cells which contained fewer pDNA complexes resulted in the highest levels of VLP stain. Finally, to determine the potential breadth of our observations, we compared daily expression patterns of ChikVLP with a reporter, monomeric GFP molecule. The similarities detected suggest the interpretations presented here to likely be more broadly informative and applicable to PEI-based transient production of additional biological products as well.
Collapse
Affiliation(s)
- Daniel J Blackstock
- Vaccine Production Program Lab, Vaccine Research Center, NIAID, NIH, Gaithersburg, MD, USA
| | - Alvenne Goh
- Vaccine Production Program Lab, Vaccine Research Center, NIAID, NIH, Gaithersburg, MD, USA
| | - Shamitha Shetty
- Vaccine Production Program Lab, Vaccine Research Center, NIAID, NIH, Gaithersburg, MD, USA
| | - Giulia Fabozzi
- Vaccine Production Program Lab, Vaccine Research Center, NIAID, NIH, Gaithersburg, MD, USA
| | - Rong Yang
- Vaccine Production Program Lab, Vaccine Research Center, NIAID, NIH, Gaithersburg, MD, USA
| | - Vera B Ivleva
- Vaccine Production Program Lab, Vaccine Research Center, NIAID, NIH, Gaithersburg, MD, USA
| | - Richard Schwartz
- Vaccine Production Program Lab, Vaccine Research Center, NIAID, NIH, Gaithersburg, MD, USA
| | - Joseph Horwitz
- Vaccine Production Program Lab, Vaccine Research Center, NIAID, NIH, Gaithersburg, MD, USA
| |
Collapse
|
24
|
Arneson-Wissink PC, Ducharme AM, Doles JD. A novel transplantable model of lung cancer-associated tissue loss and disrupted muscle regeneration. Skelet Muscle 2020; 10:6. [PMID: 32151276 PMCID: PMC7063717 DOI: 10.1186/s13395-020-00225-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/25/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Cancer-associated muscle wasting (CAW), a symptom of cancer cachexia, is associated with approximately 20% of lung cancer deaths and remains poorly characterized on a mechanistic level. Current animal models for lung cancer-associated cachexia are limited in that they (1) primarily employ flank transplantation methods, (2) have short survival times not reflective of the patient condition, and (3) are typically performed in young mice not representative of mean patient age. This study investigates a new model for lung cancer-associated cachexia that can address these issues and also implicates muscle regeneration as a contributor to CAW. METHODS We used tail vein injection as a method to introduce tumor cells that seed primarily in the lungs of mice. Body composition of tumor-bearing mice was longitudinally tracked using NMR-based, echo magnetic resonance imaging (echoMRI). These data were combined with histological and molecular assessments of skeletal muscle to provide a complete analysis of muscle wasting. RESULTS In this new lung CAW model, we observed (1) progressive loss in whole body weight, (2) progressive loss of lean and fat mass, (3) a circulating cytokine/inflammatory profile similar to that seen in other models of CAW, (4) histological changes associated with muscle wasting, and (5) molecular changes in muscle that implicate suppression of muscle repair/regeneration. Finally, we show that survival can be extended without lessening CAW by titrating injected cell number. CONCLUSIONS Overall, this study describes a new model of CAW that could be useful for further studies of lung cancer-associated wasting and accompanying changes in the regenerative capacity of muscle. Additionally, this model addresses many recent concerns with existing models such as immunocompetence, tumor location, and survival time.
Collapse
Affiliation(s)
| | - Alexandra M Ducharme
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jason D Doles
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| |
Collapse
|
25
|
Altshuler DB, Kadiyala P, Núñez FJ, Núñez FM, Carney S, Alghamri MS, Garcia-Fabiani MB, Asad AS, Nicola Candia AJ, Candolfi M, Lahann J, Moon JJ, Schwendeman A, Lowenstein PR, Castro MG. Prospects of biological and synthetic pharmacotherapies for glioblastoma. Expert Opin Biol Ther 2020; 20:305-317. [PMID: 31959027 PMCID: PMC7059118 DOI: 10.1080/14712598.2020.1713085] [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: 06/18/2019] [Accepted: 01/06/2020] [Indexed: 01/05/2023]
Abstract
Introduction: The field of neuro-oncology has experienced significant advances in recent years. More is known now about the molecular and genetic characteristics of glioma than ever before. This knowledge leads to the understanding of glioma biology and pathogenesis, guiding the development of targeted therapeutics and clinical trials. The goal of this review is to describe the state of basic, translational, and clinical research as it pertains to biological and synthetic pharmacotherapy for gliomas.Areas covered: Challenges remain in designing accurate preclinical models and identifying patients that are likely to respond to a particular targeted therapy. Preclinical models for therapeutic assessment are critical to identify the most promising treatment approaches.Expert opinion: Despite promising new therapeutics, there have been no significant breakthroughs in glioma treatment and patient outcomes. Thus, there is an urgent need to better understand the mechanisms of treatment resistance and to design effective clinical trials.
Collapse
Affiliation(s)
- David B. Altshuler
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Padma Kadiyala
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Felipe J. Núñez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Fernando M. Núñez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Stephen Carney
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Mahmoud S. Alghamri
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maria B. Garcia-Fabiani
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Antonela S. Asad
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires. Argentina
| | - Alejandro J. Nicola Candia
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires. Argentina
| | - Marianela Candolfi
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires. Argentina
| | - Joerg Lahann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - James J. Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Pedro R. Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maria G. Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
26
|
|
27
|
Wang Y, Liu S, Zhang T, Cong H, Wei Y, Xu J, Ho YP, Kong SK, Ho HP. A centrifugal microfluidic pressure regulator scheme for continuous concentration control in droplet-based microreactors. LAB ON A CHIP 2019; 19:3870-3879. [PMID: 31638632 DOI: 10.1039/c9lc00631a] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Droplet microfluidics is an emerging tool in many biological and chemical application areas such as digital polymerase chain reaction (PCR) and in vitro diagnosis because of its extremely small sample volume and wide range of possibilities for on-demand adjustment of droplet properties. Although centrifugal microfluidics has been reported as a viable scheme for droplet generation, there is not much progress as far as droplet manipulation and droplet-based reactions are concerned. In this paper, we report a microfluidic pressure regulator scheme along with the use of microcapillaries for periodic droplet generation and the subsequent fusion. This scheme enables fine control over droplet generation and the fusion process by varying the rotational frequency. To control the solution concentration in droplets, we have implemented several fusion devices, including one-to-one mode using a symmetric structure and ratio-adjustable mode with an asymmetric structure. As an application example, we performed cell transfection using the reported droplet-based technique, which resulted in considerable improvement in terms of transfection efficiency compared to the traditional bulk approach. In another example, we synthesized quasi-2D perovskites with controllable compositions and tunable photoluminescence peaks, thus confirming the volumetric accuracy of this approach down to the nano-liter scale. Compared to the common pressure pulsation approach, our centrifugal force actuation scheme offers the advantages of compactness and highly parallel batch processing. We anticipate that the new scheme will find many applications in cell biology and chemical synthesis.
Collapse
Affiliation(s)
- Yuye Wang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Shiyue Liu
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Tiankai Zhang
- Department of Electronic Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Hengji Cong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Yuanyuan Wei
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Jianbin Xu
- Department of Electronic Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Yi-Ping Ho
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Siu-Kai Kong
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Ho-Pui Ho
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| |
Collapse
|
28
|
Zhang H, Wang J, Hu M, Li BC, Li H, Chen TT, Ren KF, Ji J, Jing QM, Fu GS. Photothermal-assisted surface-mediated gene delivery for enhancing transfection efficiency. Biomater Sci 2019; 7:5177-5186. [PMID: 31588463 DOI: 10.1039/c9bm01284b] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The development of gene therapy puts forward the requirements for efficient delivery of genetic information into diverse cells. However, in some cases of transfection, especially those for transfecting some primary cells and for delivering large size plasmid DNA (pDNA), the existing conventional transfection methods show poor efficiency. How to further improve transfection efficiency in these hard-to-achieve issues remains a crucial challenge. Here, we report a photothermal-assisted surface-mediated gene delivery based on a polydopamine-polyethylenimine (PDA-PEI) surface. The PDA-PEI surface was prepared through PEI-accelerated dopamine polymerization, which showed efficiency in the immobilization of PEI/pDNA polyplexes and remarkable photothermal properties. Upon IR irradiation, we observed improved transfection efficiencies of two important hard-to-achieve transfection issues, namely the transfection of primary endothelial cells, which are kinds of typical hard-to-transfect cells, and the transfection of cells with large-size pDNA. We demonstrate that the increases of transfection efficiency were due to the hyperthermia-induced pDNA release, the local cell membrane disturbance, and the polyplex internalization. This work highlights the importance of local immobilization and release of pDNA to gene deliveries, showing great potential applications in medical devices in the field of gene therapy.
Collapse
Affiliation(s)
- He Zhang
- Department of Cardiology, Sir Run Run Shaw Hospital, MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Jing Wang
- Department of Cardiology, Sir Run Run Shaw Hospital, MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Mi Hu
- Department of Cardiology, Sir Run Run Shaw Hospital, MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Bo-Chao Li
- Department of Cardiology, Sir Run Run Shaw Hospital, MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Huan Li
- Department of Cardiology, Sir Run Run Shaw Hospital, MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Ting-Ting Chen
- Department of Cardiology, Sir Run Run Shaw Hospital, MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Ke-Feng Ren
- Department of Cardiology, Sir Run Run Shaw Hospital, MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Jian Ji
- Department of Cardiology, Sir Run Run Shaw Hospital, MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Quan-Min Jing
- General Hospital of Northern Theater Command, Shenyang 110004, China.
| | - Guo-Sheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| |
Collapse
|
29
|
Abstract
A resurgence in the development of newer gene therapy systems has led to recent successes in the treatment of B cell cancers, retinal degeneration and neuromuscular atrophy. Gene therapy offers the ability to treat the patient at the root cause of their malady by restoring normal gene function and arresting the pathological progression of their genetic disease. The current standard of care for most genetic diseases is based upon the symptomatic treatment with polypharmacy while minimizing any potential adverse effects attributed to the off-target and drug-drug interactions on the target or other organs. In the kidney, however, the development of gene therapy modifications to specific renal cells has lagged far behind those in other organ systems. Some positive strides in the past few years provide continued enthusiasm to invest the time and effort in the development of new gene therapy vectors for medical intervention to treat kidney diseases. This mini-review will systematically describe the pros and cons of the most commonly tested gene therapy vector systems derived from adenovirus, retrovirus, and adeno-associated virus and provide insight about their potential utility as a therapy for various types of genetic diseases in the kidney.
Collapse
Affiliation(s)
- Lori Davis
- College of Pharmacy, Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Frank Park
- College of Pharmacy, Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
30
|
Fu X, Chen WC, Argento C, Clarner P, Bhatt V, Dickerson R, Bou-Assaf G, Bakhshayeshi M, Lu X, Bergelson S, Pieracci J. Analytical Strategies for Quantification of Adeno-Associated Virus Empty Capsids to Support Process Development. Hum Gene Ther Methods 2019; 30:144-152. [DOI: 10.1089/hgtb.2019.088] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Xiaotong Fu
- Gene Therapy, Process Development and Biogen, Cambridge, Massachusetts
| | | | | | - Peter Clarner
- Analytical Development, Biogen, Cambridge, Massachusetts
| | - Vinay Bhatt
- Analytical Development, Biogen, Cambridge, Massachusetts
| | - Ryan Dickerson
- Gene Therapy, Process Development and Biogen, Cambridge, Massachusetts
| | | | | | - Xiaohui Lu
- Analytical Development, Biogen, Cambridge, Massachusetts
| | | | - John Pieracci
- Gene Therapy, Process Development and Biogen, Cambridge, Massachusetts
| |
Collapse
|
31
|
Lu D, Thum T. RNA-based diagnostic and therapeutic strategies for cardiovascular disease. Nat Rev Cardiol 2019; 16:661-674. [PMID: 31186539 DOI: 10.1038/s41569-019-0218-x] [Citation(s) in RCA: 231] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/03/2019] [Indexed: 12/17/2022]
Abstract
Cardiovascular diseases are the leading cause of death globally and are associated with increasing financial expenditure. With the availability of next-generation sequencing technologies since the early 2000s, non-coding RNAs such as microRNAs, long non-coding RNAs and circular RNAs have been assessed as potential therapeutic targets for numerous diseases, including cardiovascular diseases. In this Review, we summarize current approaches employed to screen for novel coding and non-coding RNA candidates with diagnostic and therapeutic potential in cardiovascular disease, including next-generation sequencing, functional high-throughput RNA screening and single-cell sequencing technologies. Furthermore, we highlight viral-based delivery tools that have been widely used to evaluate the therapeutic utility of both coding and non-coding RNAs in the context of cardiovascular disease. Finally, we discuss the potential of using oligonucleotide-based molecular products such as modified RNA, small interfering RNA and RNA mimics/inhibitors for the treatment of cardiovascular diseases. Given that many non-coding RNAs have not yet been functionally annotated, the number of potential RNA diagnostic and therapeutic targets for cardiovascular diseases will continue to expand for years to come.
Collapse
Affiliation(s)
- Dongchao Lu
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany. .,Cardior Pharmaceuticals GmbH, Hannover Medical School, Hannover, Germany. .,National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
32
|
Liu L, Ma J, Qin L, Shi X, Si H, Wei Y. Interleukin-24 enhancing antitumor activity of chimeric oncolytic adenovirus for treating acute promyelocytic leukemia cell. Medicine (Baltimore) 2019; 98:e15875. [PMID: 31145345 PMCID: PMC6708966 DOI: 10.1097/md.0000000000015875] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Acute promyelocytic leukaemia (APL) is a clonal disease arising by hematopoietic stem cell (HSC), which characterized by inappropriate proliferation/differentiation or survival of immature myeloid progenitors. Oncolytic adenoviruses have been under widespread investigation as anticancer agents. Recently, our data suggested that tumor cells were cured by AdCN205-IL-24, an adenovirus serotype 5-based conditionally replicating adenovirus expressing IL-24 after infection. METHODS In this study, we created a novel fiber chimeric oncolytic adenovirus AdCN306-IL-24 that has Ad11 tropism and approved CAR (coxsackie adenovirus receptor, CAR)-independent cell entry, which could allow development of selective cytopathic effects (CPE) in APL cells in vitro. RESULTS Formidable cytotoxic effect was specifically implemented in APL cells after infection with AdCN306-IL-24. The expression of IL-24 was up-regulated upon treated with accepted tumors. And the vector also induced superior cytolytic effects activity in APL cells by activation of programmed cell death. CONCLUSIONS Taken together, our data suggested that chimeric oncolytic adenovirus AdCN306-IL-24 could express IL-24 gene, representing a potential therapeutics for acute promyelocytic leukemia.
Collapse
Affiliation(s)
- Li Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Department of Life Science, Northwest University, Xi’an, Shannxi
- School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, P.R. China
| | - Jiabin Ma
- School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, P.R. China
| | - Lanyi Qin
- School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, P.R. China
| | - Xiaogang Shi
- School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, P.R. China
| | - Hongqiang Si
- School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, P.R. China
| | - Yahui Wei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Department of Life Science, Northwest University, Xi’an, Shannxi
| |
Collapse
|
33
|
von der Haar K, Jonczyk R, Lavrentieva A, Weyand B, Vogt P, Jochums A, Stahl F, Scheper T, Blume CA. Electroporation: A Sustainable and Cell Biology Preserving Cell Labeling Method for Adipogenous Mesenchymal Stem Cells. Biores Open Access 2019; 8:32-44. [PMID: 30944770 PMCID: PMC6445215 DOI: 10.1089/biores.2019.0001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Human mesenchymal stem cells derived from adipose tissue (AD-hMSCs) represent a promising source for tissue engineering and are already widely used in cell therapeutic clinical trials. Until today, an efficient and sustainable cell labeling system for cell tracking does not exist. We evaluated transient transfection through electroporation for cell labeling and compared it with lentiviral transduction for AD-hMSCs. In addition, we tested whether nonsense DNA or a reporter gene such as enhanced green fluorescent protein (EGFP) is the more suitable label for AD-hMSCs. Using electroporation, the transfection efficiency reached a maximal level of 44.6 ± 1.1% EGFP-positive cells after selective and expansive cultivation of the mixed MSC population, and was 44.5 ± 1.4% after gene transfer with Cyanin3-marked nonsense-label DNA, which remained stable during 2 weeks of nonselective cultivation (37.2 ± 4.7% positive AD-hMSCs). Electroporation with both nonsense DNA and pEGFP-N1 led to a slight growth retardation of 45.2% and 59.1%, respectively. EGFP-transfected or transduced AD-hMSCs showed a limited adipogenic and osteogenic differentiation capacity, whereas it was almost unaffected in cells electroporated with the nonsense-label DNA. The nonsense DNA was detectable through quantitative real-time polymerase chain reaction for at least 5 weeks/10 passages and in differentiated AD-hMSCs. EGFP-labeled cells were trackable for 24 h in vitro and served as testing cells with new materials for dental implants for 7 days. In contrast, lentivirally transduced AD-hMSCs showed an altered natural immune phenotype of the AD-hMSCs with lowered expression of two cell type defining surface markers (CD44 and CD73) and a relevantly decreased cell growth by 71.8% as assessed by the number of colony-forming units. We suggest electroporation with nonsense DNA as an efficient and long-lasting labeling method for AD-hMSCs with the comparably lowest negative impact on the phenotype or the differentiation capacity of the cells, which may, therefore, be suitable for tissue engineering. In contrast, EGFP transfection by electroporation is efficient but may be more suitable for cell tracking within cell therapies without MSC differentiation procedures. Since current protocols of lentiviral gene transduction include the risk of cell biological alterations, electroporation seems advantageous and sustainable enough for hMSC labeling.
Collapse
Affiliation(s)
- Kathrin von der Haar
- Institute of Technical Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Rebecca Jonczyk
- Institute of Technical Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Antonina Lavrentieva
- Institute of Technical Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Birgit Weyand
- Department of Plastic Hand and Reconstructive Surgery, Hannover Medical School Hannover, Hannover, Germany
| | - Peter Vogt
- Department of Plastic Hand and Reconstructive Surgery, Hannover Medical School Hannover, Hannover, Germany
| | - André Jochums
- Institute of Technical Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Frank Stahl
- Institute of Technical Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Thomas Scheper
- Institute of Technical Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Cornelia A. Blume
- Institute of Technical Chemistry, Leibniz University Hannover, Hannover, Germany
| |
Collapse
|
34
|
Vaijayanthi T, Pandian GN, Sugiyama H. Chemical Control System of Epigenetics. CHEM REC 2018; 18:1833-1853. [DOI: 10.1002/tcr.201800067] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 10/07/2018] [Indexed: 12/28/2022]
Affiliation(s)
- Thangavel Vaijayanthi
- Department of ChemistryGraduate School of ScienceKyoto University Kitashirakawa-Oiwakecho, Sakyo-ku Kyoto 606-8502, Japan
| | - Ganesh N. Pandian
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS)Kyoto University Yoshida-Ushinomaecho, Sakyo-ku Kyoto 606-8501 Japan
| | - Hiroshi Sugiyama
- Department of ChemistryGraduate School of ScienceKyoto University Kitashirakawa-Oiwakecho, Sakyo-ku Kyoto 606-8502, Japan
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS)Kyoto University Yoshida-Ushinomaecho, Sakyo-ku Kyoto 606-8501 Japan
| |
Collapse
|
35
|
Yoshimura M, Ueta Y. Advanced genetic and viral methods for labelling and manipulation of oxytocin and vasopressin neurones in rats. Cell Tissue Res 2018; 375:311-327. [PMID: 30338378 DOI: 10.1007/s00441-018-2932-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 09/19/2018] [Indexed: 12/22/2022]
Abstract
Rats have been widely used as one of the most common laboratory animals for biological research, because their physiology, pathology, and behavioral characteristics are highly similar to humans. Recent developments in rat genetic modification techniques have now led to further their utility for a broad range of research questions, including the ability to specifically label individual neurones, and even manipulate neuronal function in rats. We have succeeded in generating several transgenic rat lines that enable visualization of specific neurones due to their expression of fluorescently-tagged oxytocin, vasopressin, and c-fos protein. Furthermore, we have been able to generate novel transgenic rat lines in which we can activate vasopressin neurones using optogenetic and chemogenetic techniques. In this review, we will summarize the techniques of genetic modification for labeling and manipulating the specific neurones. Successful examples of generating transgenic rat lines in our lab and usefulness of these rats will also be introduced. These transgenic rat lines enable the interrogation of neuronal function and physiology in a way that was not possible in the past, providing novel insights into neuronal mechanisms both in vivo and ex vivo.
Collapse
Affiliation(s)
- Mitsuhiro Yoshimura
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Yoichi Ueta
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan.
| |
Collapse
|
36
|
Enhanced anticancer effect of MAP30–S3 by cyclosproin A through endosomal escape. Anticancer Drugs 2018; 29:736-747. [DOI: 10.1097/cad.0000000000000649] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
37
|
James EN, Van Doren E, Li C, Kaplan DL. Silk Biomaterials-Mediated miRNA Functionalized Orthopedic Devices. Tissue Eng Part A 2018; 25:12-23. [PMID: 29415631 DOI: 10.1089/ten.tea.2017.0455] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Silk-based bioresorbable medical devices, such as screws, plates, and rods, have been under investigation due to their promising properties for orthopedic repairs. Options to functionalize these new devices for enhanced control of bone regeneration would also exploit the compatible processing methods used to generate the devices. MicroRNAs are important regulators of bone maintenance and formation, and miRNA-based therapeutics have the potential to aid bone repair, utilizing a transient therapeutic approach with local bioactivity. We hypothesized that silk-based orthopedic devices could be used for the local delivery of miRNAs, using anti-sense miR-214 (AS-miR-214), to inhibit endogenous expression of osteoinductive antagonist and thereby supporting the upregulation of osteoinductive target molecules activating transcription factor 4 (ATF4) and Osterix (Osx). AS-miR-214 silk devices, prepared using surface coating, demonstrated continuous release of miRNA inhibitors up to 7 days in vitro. Additionally, human mesenchymal stem cells seeded on AS-miR-214 silk films expressed higher levels of osteogenic genes ATF4, Osx, Runx2, and Osteocalcin. Interestingly, these cells exhibited lower cell viability and DNA content over 21 days. Conversely, the cells demonstrated significantly higher levels of alkaline phosphatase expression and calcium deposition compared with cells seeded on silk films with nontargeting miRNA controls. The study demonstrated that the silk-based orthopedic devices, in conjunction with bioactive miRNA-based therapeutics, may serve as a novel system for localized bone tissue engineering, enhancing osteogenesis at the implant interface while avoiding detrimental systematic side effects.
Collapse
Affiliation(s)
- Eric N James
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Emily Van Doren
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Chunmei Li
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| |
Collapse
|
38
|
Polesskaya O, Baranova A, Bui S, Kondratev N, Kananykhina E, Nazarenko O, Shapiro T, Nardia FB, Kornienko V, Chandhoke V, Stadler I, Lanzafame R, Myakishev-Rempel M. Optogenetic regulation of transcription. BMC Neurosci 2018; 19:12. [PMID: 29745855 PMCID: PMC5998900 DOI: 10.1186/s12868-018-0411-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Optogenetics has become widely recognized for its success in real-time control of brain neurons by utilizing non-mammalian photosensitive proteins to open or close membrane channels. Here we review a less well known type of optogenetic constructs that employs photosensitive proteins to transduce the signal to regulate gene transcription, and its possible use in medicine. One of the problems with existing gene therapies is that they could remain active indefinitely while not allowing regulated transgene production on demand. Optogenetic regulation of transcription (ORT) could potentially be used to regulate the production of a biological drug in situ, by repeatedly applying light to the tissue, and inducing expression of therapeutic transgenes when needed. Red and near infrared wavelengths, which are capable of penetration into tissues, have potential for therapeutic applications. Existing ORT systems are reviewed herein with these considerations in mind.
Collapse
Affiliation(s)
| | - Ancha Baranova
- Research Center for Medical Genetics RAMS, Moscow, Russia.,Center for the Study of Chronic Metabolic and Rare Diseases, School of Systems Biology, George Mason University, Fairfax, VA, USA.,Atlas Biomed Group, Moscow, Russia.,Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Sarah Bui
- Center for the Study of Chronic Metabolic and Rare Diseases, School of Systems Biology, George Mason University, Fairfax, VA, USA
| | | | | | | | | | | | | | - Vikas Chandhoke
- Center for the Study of Chronic Metabolic and Rare Diseases, School of Systems Biology, George Mason University, Fairfax, VA, USA
| | | | | | - Max Myakishev-Rempel
- Localized Therapeutics, LLC, San Diego, CA, USA. .,Vaccine Research Institute of San Diego, San Diego, CA, USA.
| |
Collapse
|
39
|
López-Marín LM, Rivera AL, Fernández F, Loske AM. Shock wave-induced permeabilization of mammalian cells. Phys Life Rev 2018; 26-27:1-38. [PMID: 29685859 DOI: 10.1016/j.plrev.2018.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/12/2018] [Accepted: 02/26/2018] [Indexed: 12/18/2022]
Abstract
Controlled permeabilization of mammalian cell membranes is fundamental to develop gene and cell therapies based on macromolecular cargo delivery, a process that emerged against an increasing number of health afflictions, including genetic disorders, cancer and infections. Viral vectors have been successfully used for macromolecular delivery; however, they may have unpredictable side effects and have been limited to life-threatening cases. Thus, several chemical and physical methods have been explored to introduce drugs, vaccines, and nucleic acids into cells. One of the most appealing physical methods to deliver genes into cells is shock wave-induced poration. High-speed microjets of fluid, emitted due to the collapse of microbubbles after shock wave passage, represent the most significant mechanism that contributes to cell membrane poration by this technique. Herein, progress in shock wave-induced permeabilization of mammalian cells is presented. After covering the main concepts related to molecular strategies whose applications depend on safer drug delivery methods, the physics behind shock wave phenomena is described. Insights into the use of shock waves for cell membrane permeation are discussed, along with an overview of the two major biomedical applications thereof-i.e., genetic modification and anti-cancer shock wave-assisted chemotherapy. The aim of this review is to summarize 30 years of data showing underwater shock waves as a safe, noninvasive method for macromolecular delivery into mammalian cells, encouraging the development of further research, which is still required before the introduction of this promising tool into clinical practice.
Collapse
Affiliation(s)
- Luz M López-Marín
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, 76230 Querétaro, Qro., Mexico.
| | - Ana Leonor Rivera
- Instituto de Ciencias Nucleares & Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510 Ciudad de México, Mexico.
| | - Francisco Fernández
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, 76230 Querétaro, Qro., Mexico.
| | - Achim M Loske
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, 76230 Querétaro, Qro., Mexico.
| |
Collapse
|
40
|
Yang Y, Hong Y, Cho E, Kim GB, Kim IS. Extracellular vesicles as a platform for membrane-associated therapeutic protein delivery. J Extracell Vesicles 2018; 7:1440131. [PMID: 29535849 PMCID: PMC5844050 DOI: 10.1080/20013078.2018.1440131] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 02/07/2018] [Indexed: 02/08/2023] Open
Abstract
Membrane proteins are of great research interest, particularly because they are rich in targets for therapeutic application. The suitability of various membrane proteins as targets for therapeutic formulations, such as drugs or antibodies, has been studied in preclinical and clinical studies. For therapeutic application, however, a protein must be expressed and purified in as close to its native conformation as possible. This has proven difficult for membrane proteins, as their native conformation requires the association with an appropriate cellular membrane. One solution to this problem is to use extracellular vesicles as a display platform. Exosomes and microvesicles are membranous extracellular vesicles that are released from most cells. Their membranes may provide a favourable microenvironment for membrane proteins to take on their proper conformation, activity, and membrane distribution; moreover, membrane proteins can cluster into microdomains on the surface of extracellular vesicles following their biogenesis. In this review, we survey the state-of-the-art of extracellular vesicle (exosome and small-sized microvesicle)-based therapeutics, evaluate the current biological understanding of these formulations, and forecast the technical advances that will be needed to continue driving the development of membrane protein therapeutics.
Collapse
Affiliation(s)
- Yoosoo Yang
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division for Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
| | - Yeonsun Hong
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Eunji Cho
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Gi Beom Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - In-San Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| |
Collapse
|
41
|
Terashima T, Ogawa N, Nakae Y, Sato T, Katagi M, Okano J, Maegawa H, Kojima H. Gene Therapy for Neuropathic Pain through siRNA-IRF5 Gene Delivery with Homing Peptides to Microglia. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 11:203-215. [PMID: 29858055 PMCID: PMC5992689 DOI: 10.1016/j.omtn.2018.02.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 02/02/2018] [Accepted: 02/21/2018] [Indexed: 01/01/2023]
Abstract
Astrocyte- and microglia-targeting peptides were identified and isolated using phage display technology. A series of procedures, including three cycles of both in vivo and in vitro biopanning, was performed separately in astrocytes and in M1 or M2 microglia, yielding 50–58 phage plaques in each cell type. Analyses of the sequences of this collection identified one candidate homing peptide targeting astrocytes (AS1[C-LNSSQPS-C]) and two candidate homing peptides targeting microglia (MG1[C-HHSSSAR-C] and MG2[C-NTGSPYE-C]). To determine peptide specificity for the target cell in vitro, each peptide was synthesized and introduced into the primary cultures of astrocytes or microglia. Those peptides could bind to the target cells and be selectively taken up by the corresponding cell, namely, astrocytes, M1 microglia, or M2 microglia. To confirm cell-specific gene delivery to M1 microglia, the complexes between peptide MG1 and siRNA-interferon regulatory factor 5 were prepared and intrathecally injected into a mouse model of neuropathic pain. The complexes successfully suppressed hyperalgesia with high efficiency in this neuropathic pain model. Here, we describe a novel gene therapy for the treatment neuropathic pain, which has a high potential to be of clinical relevance. This strategy will ensure the targeted delivery of therapeutic genes while minimizing side effects to non-target tissues or cells.
Collapse
Affiliation(s)
- Tomoya Terashima
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Shiga, Japan.
| | - Nobuhiro Ogawa
- Department of Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Yuki Nakae
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Toshiyuki Sato
- Pain & Neuroscience Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Miwako Katagi
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Junko Okano
- Division of Anatomy and Cell Biology, Shiga University of Medical Science, Shiga, Japan
| | - Hiroshi Maegawa
- Department of Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Hideto Kojima
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Shiga, Japan
| |
Collapse
|
42
|
Hu WW, Yeh CC, Tsai CW. The conjugation of indolicidin to polyethylenimine for enhanced gene delivery with reduced cytotoxicity. J Mater Chem B 2018; 6:5781-5794. [DOI: 10.1039/c8tb01408f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The hydrophobic domains of conjugated peptides can stabilize their insertion into the cell membrane to promote transportation.
Collapse
Affiliation(s)
- Wei-Wen Hu
- Department of Chemical and Materials Engineering
- National Central University
- Taoyuan City
- Taiwan
- Center for Biocellular Engineering
| | - Chiao-Chun Yeh
- Department of Chemical and Materials Engineering
- National Central University
- Taoyuan City
- Taiwan
| | - Ching-Wei Tsai
- Department of Chemical and Materials Engineering
- National Central University
- Taoyuan City
- Taiwan
| |
Collapse
|
43
|
Abstract
Green Fluorescent protein (GFP), used as a cellular tag, provides researchers with a valuable method of measuring gene expression and cell tracking. However, there is evidence to suggest that the immunogenicity and cytotoxicity of GFP potentially confounds the interpretation of in vivo experimental data. Studies have shown that GFP expression can deteriorate over time as GFP tagged cells are prone to death. Therefore, the cells that were originally marked with GFP do not survive and cannot be accurately traced over time. This review will present current evidence for the immunogenicity and cytotoxicity of GFP in in vivo studies by characterizing these responses.
Collapse
|
44
|
Süleymanoğlu E. Mg 2+-induced DNA compaction, condensation, and phase separation in gene delivery vehicles based on zwitterionic phospholipids: a dynamic light scattering and surface-enhanced Raman spectroscopic study. J Biol Inorg Chem 2017; 22:1165-1177. [PMID: 28924921 DOI: 10.1007/s00775-017-1492-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 08/28/2017] [Indexed: 01/08/2023]
Abstract
Despite the significant efforts towards applying improved non-destructive and label-free measurements of biomolecular structures of lipid-based gene delivery vectors, little is achieved in terms of their structural relevance in gene transfections. Better understanding of structure-activity relationships of lipid-DNA complexes and their gene expression efficiencies thus becomes an essential issue. Raman scattering offers a complimentary measurement technique for following the structural transitions of both DNA and lipid vesicles employed for their transfer. This work describes the use of SERS coupled with light scattering approaches for deciphering the bioelectrochemical phase formations between nucleic acids and lipid vesicles within lipoplexes and their surface parameters that could influence both the uptake of non-viral gene carriers and the endocytic routes of interacting cells. As promising non-viral alternatives of currently employed risky viral systems or highly cytotoxic cationic liposomes, complexations of both nucleic acids and zwitterionic lipids in the presence of Mg2+ were studied applying colloidal Ag nanoparticles. It is shown that the results could be employed in further conformational characterizations of similar polyelectrolyte gene delivery systems.
Collapse
Affiliation(s)
- Erhan Süleymanoğlu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, 06330, Ankara, Turkey.
| |
Collapse
|
45
|
Bhounsule AS, Bhatt LK, Prabhavalkar KS, Oza M. Cyclin dependent kinase 5: A novel avenue for Alzheimer’s disease. Brain Res Bull 2017; 132:28-38. [DOI: 10.1016/j.brainresbull.2017.05.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 05/10/2017] [Indexed: 10/19/2022]
|
46
|
Rezaie J, Ajezi S, Avci ÇB, Karimipour M, Geranmayeh MH, Nourazarian A, Sokullu E, Rezabakhsh A, Rahbarghazi R. Exosomes and their Application in Biomedical Field: Difficulties and Advantages. Mol Neurobiol 2017; 55:3372-3393. [DOI: 10.1007/s12035-017-0582-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/27/2017] [Indexed: 12/31/2022]
|
47
|
Ni R, Zhou J, Hossain N, Chau Y. Virus-inspired nucleic acid delivery system: Linking virus and viral mimicry. Adv Drug Deliv Rev 2016; 106:3-26. [PMID: 27473931 DOI: 10.1016/j.addr.2016.07.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 07/02/2016] [Accepted: 07/20/2016] [Indexed: 12/21/2022]
Abstract
Targeted delivery of nucleic acids into disease sites of human body has been attempted for decades, but both viral and non-viral vectors are yet to meet our expectations. Safety concerns and low delivery efficiency are the main limitations of viral and non-viral vectors, respectively. The structure of viruses is both ordered and dynamic, and is believed to be the key for effective transfection. Detailed understanding of the physical properties of viruses, their interaction with cellular components, and responses towards cellular environments leading to transfection would inspire the development of safe and effective non-viral vectors. To this goal, this review systematically summarizes distinctive features of viruses that are implied for efficient nucleic acid delivery but not yet fully explored in current non-viral vectors. The assembly and disassembly of viral structures, presentation of viral ligands, and the subcellular targeting of viruses are emphasized. Moreover, we describe the current development of cationic material-based viral mimicry (CVM) and structural viral mimicry (SVM) in these aspects. In light of the discrepancy, we identify future opportunities for rational design of viral mimics for the efficient delivery of DNA and RNA.
Collapse
Affiliation(s)
- Rong Ni
- Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Institute for Advanced Study, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Junli Zhou
- Department of Chemical and Biomolecular Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Naushad Hossain
- Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Ying Chau
- Department of Chemical and Biomolecular Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.
| |
Collapse
|
48
|
Isik M, Agirre M, Zarate J, Puras G, Mecerreyes D, Sardon H, Pedraz JL. Amine containing cationic methacrylate copolymers as efficient gene delivery vehicles to retinal epithelial cells. ACTA ACUST UNITED AC 2016. [DOI: 10.1002/pola.28376] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Mehmet Isik
- POLYMAT University of the Basque Country UPV/EHU, Joxe Mari Korta Center; Avda. Tolosa 72 Donostia-San Sebastian 20018 Spain
| | - Mireia Agirre
- NanoBioCel Group, University of the Basque Country UPV/EHU; Vitoria-Gasteiz Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN); Vitoria-Gasteiz Spain
| | - Jon Zarate
- NanoBioCel Group, University of the Basque Country UPV/EHU; Vitoria-Gasteiz Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN); Vitoria-Gasteiz Spain
| | - Gustavo Puras
- NanoBioCel Group, University of the Basque Country UPV/EHU; Vitoria-Gasteiz Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN); Vitoria-Gasteiz Spain
| | - David Mecerreyes
- POLYMAT University of the Basque Country UPV/EHU, Joxe Mari Korta Center; Avda. Tolosa 72 Donostia-San Sebastian 20018 Spain
- Basque Foundation for Science; Ikerbasque; Bilbao E-48011 Spain
| | - Haritz Sardon
- POLYMAT University of the Basque Country UPV/EHU, Joxe Mari Korta Center; Avda. Tolosa 72 Donostia-San Sebastian 20018 Spain
| | - J. L. Pedraz
- NanoBioCel Group, University of the Basque Country UPV/EHU; Vitoria-Gasteiz Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN); Vitoria-Gasteiz Spain
| |
Collapse
|
49
|
Vosen S, Rieck S, Heidsieck A, Mykhaylyk O, Zimmermann K, Plank C, Gleich B, Pfeifer A, Fleischmann BK, Wenzel D. Improvement of vascular function by magnetic nanoparticle-assisted circumferential gene transfer into the native endothelium. J Control Release 2016; 241:164-173. [PMID: 27667178 DOI: 10.1016/j.jconrel.2016.09.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/20/2016] [Accepted: 09/21/2016] [Indexed: 12/22/2022]
Abstract
Gene therapy is a promising approach for chronic disorders that require continuous treatment such as cardiovascular disease. Overexpression of vasoprotective genes has generated encouraging results in animal models, but not in clinical trials. One major problem in humans is the delivery of sufficient amounts of genetic vectors to the endothelium which is impeded by blood flow, whereas prolonged stop-flow conditions impose the risk of ischemia. In the current study we have therefore developed a strategy for the efficient circumferential lentiviral gene transfer in the native endothelium under constant flow conditions. For that purpose we perfused vessels that were exposed to specially designed magnetic fields with complexes of lentivirus and magnetic nanoparticles thereby enabling overexpression of therapeutic genes such as endothelial nitric oxide synthase (eNOS) and vascular endothelial growth factor (VEGF). This treatment enhanced NO and VEGF production in the transduced endothelium and resulted in a reduction of vascular tone and increased angiogenesis. Thus, the combination of MNPs with magnetic fields is an innovative strategy for site-specific and efficient vascular gene therapy.
Collapse
Affiliation(s)
- Sarah Vosen
- Institute of Physiology I, Life & Brain Center, University Clinic of Bonn, Germany
| | - Sarah Rieck
- Institute of Physiology I, Life & Brain Center, University Clinic of Bonn, Germany
| | | | - Olga Mykhaylyk
- Institute of Experimental Oncology and Therapy Research, TU München, Germany
| | - Katrin Zimmermann
- Institute of Pharmacology and Toxicology, University Clinic of Bonn, Germany
| | - Christian Plank
- Institute of Experimental Oncology and Therapy Research, TU München, Germany
| | - Bernhard Gleich
- Zentralinstitut für Medizintechnik (IMETUM), TU München, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Clinic of Bonn, Germany
| | - Bernd K Fleischmann
- Institute of Physiology I, Life & Brain Center, University Clinic of Bonn, Germany
| | - Daniela Wenzel
- Institute of Physiology I, Life & Brain Center, University Clinic of Bonn, Germany.
| |
Collapse
|
50
|
Balkow A, Hoffmann LS, Klepac K, Glöde A, Gnad T, Zimmermann K, Pfeifer A. Direct lentivirus injection for fast and efficient gene transfer into brown and beige adipose tissue. J Biol Methods 2016; 3:e48. [PMID: 31453213 PMCID: PMC6706150 DOI: 10.14440/jbm.2016.123] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 05/23/2016] [Accepted: 06/27/2016] [Indexed: 11/23/2022] Open
Abstract
Brown adipose tissue is a special type of fat contributing to energy expenditure in human newborns and adults. Moreover, subcutaneous white adipose tissue has a high capacity to adapt an energy-consuming, brown-like/beige phenotype. Here, we developed an easy to handle and fast to accomplish method to efficiently transfer genes into brown and beige fat pads in vivo. Lentiviral vectors are directly injected into the target fat pad of anesthetized mice through a small incision using a modified, small needle connected to a microsyringe, which is well suited for infiltration of adipose tissues. Expression of the target gene can be detected in brown/beige fat one week after injection. The method can be applied within minutes to efficiently deliver transgenes into subcutaneous adipose tissues. Thus, this protocol allows for studying genes of interest in a timely manner in murine brown/beige fat and could potentially lead to new gene therapies for obesity.
Collapse
Affiliation(s)
- Aileen Balkow
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Linda S Hoffmann
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Katarina Klepac
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany.,Research Training Group 1873, University of Bonn, 53127 Bonn, Germany
| | - Anja Glöde
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany.,BIGS DrugS International Graduate School, University of Bonn, 53127 Bonn, Germany
| | - Thorsten Gnad
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Katrin Zimmermann
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany.,Research Training Group 1873, University of Bonn, 53127 Bonn, Germany.,BIGS DrugS International Graduate School, University of Bonn, 53127 Bonn, Germany.,PharmaCenter, University of Bonn, 53127 Bonn, Germany
| |
Collapse
|