1
|
Idiago-López J, Ferreira D, Asín L, Moros M, Armenia I, Grazú V, Fernandes AR, de la Fuente JM, Baptista PV, Fratila RM. Membrane-localized magnetic hyperthermia promotes intracellular delivery of cell-impermeant probes. NANOSCALE 2024; 16:15176-15195. [PMID: 39052238 DOI: 10.1039/d4nr01955e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
In this work, we report the disruptive use of membrane-localized magnetic hyperthermia to promote the internalization of cell-impermeant probes. Under an alternating magnetic field, magnetic nanoparticles (MNPs) immobilized on the cell membrane via bioorthogonal click chemistry act as nanoheaters and lead to the thermal disruption of the plasma membrane, which can be used for internalization of different types of molecules, such as small fluorescent probes and nucleic acids. Noteworthily, no cell death, oxidative stress and alterations of the cell cycle are detected after the thermal stimulus, although cells are able to sense and respond to the thermal stimulus through the expression of different types of heat shock proteins (HSPs). Finally, we demonstrate the utility of this approach for the transfection of cells with a small interference RNA (siRNA), revealing a similar efficacy to a standard transfection method based on the use of cationic lipid-based reagents (such as Lipofectamine), but with lower cell toxicity. These results open the possibility of developing new procedures for "opening and closing" cellular membranes with minimal disturbance of cellular integrity. This on-demand modification of cell membrane permeability could allow the direct intracellular delivery of biologically relevant (bio)molecules, drugs and nanomaterials, thus overcoming traditional endocytosis pathways and avoiding endosomal entrapment.
Collapse
Affiliation(s)
- Javier Idiago-López
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, 50009, Zaragoza, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Daniela Ferreira
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal.
- UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Laura Asín
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, 50009, Zaragoza, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - María Moros
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, 50009, Zaragoza, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Ilaria Armenia
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, 50009, Zaragoza, Spain.
| | - Valeria Grazú
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, 50009, Zaragoza, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Alexandra R Fernandes
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal.
- UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Jesús M de la Fuente
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, 50009, Zaragoza, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Pedro V Baptista
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal.
- UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Raluca M Fratila
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, 50009, Zaragoza, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
- Departamento de Química Orgánica, Facultad de Ciencias, C/Pedro Cerbuna 12, 50009, Zaragoza, Spain
| |
Collapse
|
2
|
Skinner MA, Otten A, Hoff A, Jaroszeski M. Combined effect of heat and corona charge on molecular delivery to a T cell line in vitro. PLoS One 2023; 18:e0293035. [PMID: 37851653 PMCID: PMC10584139 DOI: 10.1371/journal.pone.0293035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/03/2023] [Indexed: 10/20/2023] Open
Abstract
With the rapid increase of gene and immunotherapies for treating cancer, there is a need to efficiently transfect cells. Previous studies suggest that electrotransfer can provide a non-viral method for gene delivery. Electrotransfer traditionally relies upon the application of direct current pulses to the cells of interest. Corona charge was investigated in this study as an alternative to traditional methods as a means of creating the electric field necessary to deliver materials via electrotransfer. The goal was to determine if there was an increase in molecular delivery across the membrane of a human T cell line used as a model system. In a novel dish created for the study, the effects of elevated temperatures (37, 40, 43, and 45°C) during the treatment process were also examined in combination with corona charge application. Results showed that treating cells with corona charge at room temperature (~23°C) caused a statistically significant increase in molecular delivery while maintaining viability. Heat alone did not cause a statistically significant effect on molecular delivery. Combined corona charge treatment and heating resulted in a statistically significant increase on molecular delivery compared to controls that were only heated. Combined corona charge treatment and heating to all temperatures when compared to controls treated at room temperature, showed a statistically significant increase in molecular delivery.
Collapse
Affiliation(s)
- Molly A. Skinner
- Department of Chemical, Biological, and Materials Engineering, University of South Florida, Tampa, FL, United States of America
| | - Alex Otten
- Department of Electrical Engineering, University of South Florida, Tampa, FL, United States of America
| | - Andrew Hoff
- Department of Electrical Engineering, University of South Florida, Tampa, FL, United States of America
| | - Mark Jaroszeski
- Department of Medical Engineering University of South Florida, Tampa, FL, United States of America
| |
Collapse
|
3
|
Abstract
» Orthopaedics pioneered the expansion of gene therapy beyond its traditional scope of diseases that are caused by rare single-gene defects. Orthopaedic applications of gene therapy are most developed in the areas of arthritis and regenerative medicine, but several additional possibilities exist. » Invossa, an ex vivo gene therapeutic for osteoarthritis, was approved in South Korea in 2017, but its approval was retracted in 2019 and remains under appeal; a Phase-III clinical trial of Invossa has restarted in the U.S. » There are several additional clinical trials for osteoarthritis and rheumatoid arthritis that could lead to approved gene therapeutics for arthritis. » Bone-healing and cartilage repair are additional areas that are attracting considerable research; intervertebral disc degeneration and the healing of ligaments, tendons, and menisci are other applications of interest. Orthopaedic tumors, genetic diseases, and aseptic loosening are additional potential targets. » If successful, these endeavors will expand the scope of gene therapy from providing expensive medicines for a few patients to providing affordable medicines for many.
Collapse
|
4
|
Bonny AR, Fonseca JP, Park JE, El-Samad H. Orthogonal control of mean and variability of endogenous genes in a human cell line. Nat Commun 2021; 12:292. [PMID: 33436569 PMCID: PMC7804932 DOI: 10.1038/s41467-020-20467-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 11/25/2020] [Indexed: 12/11/2022] Open
Abstract
Stochastic fluctuations at the transcriptional level contribute to isogenic cell-to-cell heterogeneity in mammalian cell populations. However, we still have no clear understanding of the repercussions of this heterogeneity, given the lack of tools to independently control mean expression and variability of a gene. Here, we engineer a synthetic circuit to modulate mean expression and heterogeneity of transgenes and endogenous human genes. The circuit, a Tunable Noise Rheostat (TuNR), consists of a transcriptional cascade of two inducible transcriptional activators, where the output mean and variance can be modulated by two orthogonal small molecule inputs. In this fashion, different combinations of the inputs can achieve the same mean but with different population variability. With TuNR, we achieve low basal expression, over 1000-fold expression of a transgene product, and up to 7-fold induction of the endogenous gene NGFR. Importantly, for the same mean expression level, we are able to establish varying degrees of heterogeneity in expression within an isogenic population, thereby decoupling gene expression noise from its mean. TuNR is therefore a modular tool that can be used in mammalian cells to enable direct interrogation of the implications of cell-to-cell variability.
Collapse
Affiliation(s)
- Alain R Bonny
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - João Pedro Fonseca
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94158, USA
- Amyris Bio Products Portugal, Porto, Portugal
| | - Jesslyn E Park
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Hana El-Samad
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94158, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, 94158, USA.
| |
Collapse
|
5
|
Abstract
Over 450 million people worldwide suffer from hearing loss, leading to an estimated economic burden of ∼$750 billion. The past decade has seen significant advances in the understanding of the molecular mechanisms that contribute to hearing, and the environmental and genetic factors that can go awry and lead to hearing loss. This in turn has sparked enormous interest in developing gene therapy approaches to treat this disorder. This review documents the most recent advances in cochlear gene therapy to restore hearing loss, and will cover viral vectors and construct designs, potential routes of delivery into the inner ear, and, lastly, the most promising genes of interest.
Collapse
Affiliation(s)
- Lawrence Lustig
- Department of Otolaryngology-Head and Neck Surgery, Columbia University Medical Center, New York Presbyterian Hospital, New York, New York 10032
| | - Omar Akil
- Department of Otolaryngology-Head and Neck Surgery, University of California San Francisco, San Francisco, California 94117
| |
Collapse
|
6
|
Krishnan T, David AL. Placenta-directed gene therapy for fetal growth restriction. Semin Fetal Neonatal Med 2017; 22:415-422. [PMID: 28522033 DOI: 10.1016/j.siny.2017.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Fetal growth restriction (FGR) is a serious pregnancy complication affecting ∼8% of all pregnancies. There is no treatment to increase fetal growth in the uterus. Gene therapy presents a promising treatment strategy for FGR, with the use of adenoviral vectors encoding for proteins such as vascular endothelial growth factor (VEGF) and insulin-like growth factor demonstrating improvements in fetal growth, placental function, and neonatal outcome in preclinical studies. Safety assessments suggest no adverse risk to the mother or fetus for VEGF maternal gene therapy; a clinical trial is in development. This review assesses research into placenta-directed gene therapy for FGR, investigating the use of transgenes and vectors, their route of administration in obstetrics, and the steps that will be needed to take this treatment modality into the clinic.
Collapse
Affiliation(s)
- Tara Krishnan
- UCL Institute for Women's Health, University College London, London, United Kingdom.
| | - Anna L David
- Head of Research Department of Maternal Fetal Medicine at the Institute for Women's Health, University College London, United Kingdom
| |
Collapse
|
7
|
Guo ZS, Liu Z, Sathaiah M, Wang J, Ravindranathan R, Kim E, Huang S, Kenniston TW, Bell JC, Zeh HJ, Butterfield LH, Gambotto A, Bartlett DL. Rapid Generation of Multiple Loci-Engineered Marker-free Poxvirus and Characterization of a Clinical-Grade Oncolytic Vaccinia Virus. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 7:112-122. [PMID: 29085848 PMCID: PMC5651493 DOI: 10.1016/j.omtm.2017.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 09/27/2017] [Indexed: 12/13/2022]
Abstract
Recombinant poxviruses, utilized as vaccine vectors and oncolytic viruses, often require manipulation at multiple genetic loci in the viral genome. It is essential for viral vectors to possess no adventitious mutations and no (antibiotic) selection marker in the final product for human patients in order to comply with the guidance from the regulatory agencies. Rintoul et al. have previously developed a selectable and excisable marker (SEM) system for the rapid generation of recombinant vaccinia virus. In the current study, we describe an improved methodology for rapid creation and selection of recombinant poxviruses with multiple genetic manipulations solely based on expression of a fluorescent protein and with no requirement for drug selection that can lead to cellular stress and the risk of adventitious mutations throughout the viral genome. Using this improved procedure combined with the SEM system, we have constructed multiple marker-free oncolytic poxviruses expressing different cytokines and other therapeutic genes. The high fidelity of inserted DNA sequences validates the utility of this improved procedure for generation of therapeutic viruses for human patients. We have created an oncolytic poxvirus expressing human chemokine CCL5, designated as vvDD-A34R-hCCL5, with manipulations at two genetic loci in a single virus. Finally, we have produced and purified this virus in clinical grade for its use in a phase I clinical trial and presented data on initial in vitro characterization of the virus.
Collapse
Affiliation(s)
- Zong Sheng Guo
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Corresponding author: Zong Sheng Guo, PhD, UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Zuqiang Liu
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Magesh Sathaiah
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jiahu Wang
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Roshni Ravindranathan
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eun Kim
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Shaohua Huang
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Thomas W. Kenniston
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - John C. Bell
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Herbert J. Zeh
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lisa H. Butterfield
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Departments of Medicine and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Andrea Gambotto
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - David L. Bartlett
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
8
|
Hollevoet K, Declerck PJ. State of play and clinical prospects of antibody gene transfer. J Transl Med 2017; 15:131. [PMID: 28592330 PMCID: PMC5463339 DOI: 10.1186/s12967-017-1234-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 05/31/2017] [Indexed: 12/31/2022] Open
Abstract
Recombinant monoclonal antibodies (mAbs) are one of today's most successful therapeutic classes in inflammatory diseases and oncology. A wider accessibility and implementation, however, is hampered by the high product cost and prolonged need for frequent administration. The surge in more effective mAb combination therapies further adds to the costs and risk of toxicity. To address these issues, antibody gene transfer seeks to administer to patients the mAb-encoding nucleotide sequence, rather than the mAb protein. This allows the body to produce its own medicine in a cost- and labor-effective manner, for a prolonged period of time. Expressed mAbs can be secreted systemically or locally, depending on the production site. The current review outlines the state of play and clinical prospects of antibody gene transfer, thereby highlighting recent innovations, opportunities and remaining hurdles. Different expression platforms and a multitude of administration sites have been pursued. Viral vector-mediated mAb expression thereby made the most significant strides. Therapeutic proof of concept has been demonstrated in mice and non-human primates, and intramuscular vectored mAb therapy is under clinical evaluation. However, viral vectors face limitations, particularly in terms of immunogenicity. In recent years, naked DNA has gained ground as an alternative. Attained serum mAb titers in mice, however, remain far below those obtained with viral vectors, and robust pharmacokinetic data in larger animals is limited. The broad translatability of DNA-based antibody therapy remains uncertain, despite ongoing evaluation in patients. RNA presents another emerging platform for antibody gene transfer. Early reports in mice show that mRNA may be able to rival with viral vectors in terms of generated serum mAb titers, although expression appears more short-lived. Overall, substantial progress has been made in the clinical translation of antibody gene transfer. While challenges persist, clinical prospects are amplified by ongoing innovations and the versatility of antibody gene transfer. Clinical introduction can be expedited by selecting the platform approach currently best suited for the mAb or disease of interest. Innovations in expression platform, administration and antibody technology are expected to further improve overall safety and efficacy, and unlock the vast clinical potential of antibody gene transfer.
Collapse
Affiliation(s)
- Kevin Hollevoet
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven - University of Leuven, Campus Gasthuisberg O&N 2, P.B. 820, Herestraat 49, 3000 Leuven, Belgium
| | - Paul J. Declerck
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven - University of Leuven, Campus Gasthuisberg O&N 2, P.B. 820, Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
9
|
Atkins RM, Fawcett TJ, Gilbert R, Hoff AM, Connolly R, Brown DW, Llewellyn AJ, Jaroszeski MJ. Impedance spectroscopy as an indicator for successful in vivo electric field mediated gene delivery in a murine model. Bioelectrochemistry 2017; 115:33-40. [DOI: 10.1016/j.bioelechem.2017.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 01/20/2017] [Accepted: 01/22/2017] [Indexed: 12/19/2022]
|
10
|
Quaranta P, Focosi D, Freer G, Pistello M. Tweaking Mesenchymal Stem/Progenitor Cell Immunomodulatory Properties with Viral Vectors Delivering Cytokines. Stem Cells Dev 2016; 25:1321-41. [PMID: 27476883 DOI: 10.1089/scd.2016.0145] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal Stem Cells (MSCs) can be found in various body sites. Their main role is to differentiate into cartilage, bone, muscle, and fat cells to allow tissue maintenance and repair. During inflammation, MSCs exhibit important immunomodulatory properties that are not constitutive, but require activation, upon which they may exert immunosuppressive functions. MSCs are defined as "sensors of inflammation" since they modulate their ability of interfering with the immune system both in vitro and in vivo upon interaction with different factors. MSCs may influence immune responses through different mechanisms, such as direct cell-to-cell contact, release of soluble factors, and through the induction of anergy and apoptosis. Human MSCs are defined as plastic-adherent cells expressing specific surface molecules. Lack of MHC class II antigens makes them appealing as allogeneic tools for the therapy of both autoimmune diseases and cancer. MSC therapeutic potential could be highly enhanced by the expression of exogenous cytokines provided by transduction with viral vectors. In this review, we attempt to summarize the results of a great number of in vitro and in vivo studies aimed at improving the ability of MSCs as immunomodulators in the therapy of autoimmune, degenerative diseases and cancer. We will also compare results obtained with different vectors to deliver heterologous genes to these cells.
Collapse
Affiliation(s)
- Paola Quaranta
- 1 Department of Translational Research and New Technologies in Medicine and Surgery, Virology Section and Retrovirus Center, University of Pisa , Pisa, Italy
| | - Daniele Focosi
- 2 North-Western Tuscany Blood Bank, Pisa University Hospital , Pisa, Italy
| | - Giulia Freer
- 1 Department of Translational Research and New Technologies in Medicine and Surgery, Virology Section and Retrovirus Center, University of Pisa , Pisa, Italy .,3 Virology Unit, Pisa University Hospital , Pisa, Italy
| | - Mauro Pistello
- 1 Department of Translational Research and New Technologies in Medicine and Surgery, Virology Section and Retrovirus Center, University of Pisa , Pisa, Italy .,3 Virology Unit, Pisa University Hospital , Pisa, Italy
| |
Collapse
|
11
|
Rivera-Delgado E, Ward E, von Recum HA. Providing sustained transgene induction through affinity-based drug delivery. J Biomed Mater Res A 2016; 104:1135-42. [DOI: 10.1002/jbm.a.35643] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/16/2015] [Accepted: 01/07/2016] [Indexed: 11/08/2022]
Affiliation(s)
| | - Emily Ward
- Department of Biomedical Engineering; Case Western Reserve University Cleveland; Ohio
| | - Horst A. von Recum
- Department of Biomedical Engineering; Case Western Reserve University Cleveland; Ohio
| |
Collapse
|
12
|
Abstract
Nucleic acids show immense potential to treat cancer, acquired immune deficiency syndrome, neurological diseases and other incurable human diseases. Upon systemic administration, they encounter a series of barriers and hence barely reach the site of action, the cell. Intracellular delivery of nucleic acids is facilitated by nanovectors, both viral and non-viral. A major advantage of non-viral vectors over viral vectors is safety. Nanovectors evaluated specifically for nucleic acid delivery include polyplexes, lipoplexes and other cationic carrier-based vectors. However, more recently there is an increased interest in inorganic nanovectors for nucleic acid delivery. Nevertheless, there is no comprehensive review on the subject. The present review would cover in detail specific properties and types of inorganic nanovectors, their preparation techniques and various biomedical applications as therapeutics, diagnostics and theranostics. Future prospects are also suggested.
Collapse
|
13
|
Chen IY, Paulmurugan R, Nielsen CH, Wang DS, Chow V, Robbins RC, Gambhir SS. A titratable two-step transcriptional amplification strategy for targeted gene therapy based on ligand-induced intramolecular folding of a mutant human estrogen receptor. Mol Imaging Biol 2014; 16:224-34. [PMID: 23955099 DOI: 10.1007/s11307-013-0673-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE The efficacy and safety of cardiac gene therapy depend critically on the level and the distribution of therapeutic gene expression following vector administration. We aimed to develop a titratable two-step transcriptional amplification (tTSTA) vector strategy, which allows modulation of transcriptionally targeted gene expression in the myocardium. PROCEDURES We constructed a tTSTA plasmid vector (pcTnT-tTSTA-fluc), which uses the cardiac troponin T (cTnT) promoter to drive the expression of the recombinant transcriptional activator GAL4-mER(LBD)-VP2, whose ability to transactivate the downstream firefly luciferase reporter gene (fluc) depends on the binding of its mutant estrogen receptor (ER(G521T)) ligand binding domain (LBD) to an ER ligand such as raloxifene. Mice underwent either intramyocardial or hydrodynamic tail vein (HTV) injection of pcTnT-tTSTA-fluc, followed by differential modulation of fluc expression with varying doses of intraperitoneal raloxifene prior to bioluminescence imaging to assess the kinetics of myocardial or hepatic fluc expression. RESULTS Intramyocardial injection of pcTnT-tTSTA-fluc followed by titration with intraperitoneal raloxifene led to up to tenfold induction of myocardial fluc expression. HTV injection of pcTnT-tTSTA-fluc led to negligible long-term hepatic fluc expression, regardless of the raloxifene dose given. CONCLUSIONS The tTSTA vector strategy can effectively modulate transgene expression in a tissue-specific manner. Further refinement of this strategy should help maximize the benefit-to-risk ratio of cardiac gene therapy.
Collapse
Affiliation(s)
- Ian Y Chen
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Lalitha K, Muthusamy K, Prasad YS, Vemula PK, Nagarajan S. Recent developments in β-C-glycosides: synthesis and applications. Carbohydr Res 2014; 402:158-71. [PMID: 25498016 DOI: 10.1016/j.carres.2014.10.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 10/11/2014] [Accepted: 10/16/2014] [Indexed: 11/26/2022]
Abstract
In the last few years, considerable progress has been made in the synthesis of C-glycosides. Despite its challenging chemistry, due to its versatility, C-glycosides play a pivotal role in developing novel materials, surfactants and bioactive molecules. In this review, we present snapshots of various synthetic methodologies developed for C-glycosides in the recent years and the potential application of C-glycosides derived from β-C-glycosidic ketones.
Collapse
Affiliation(s)
- Krishnamoorthy Lalitha
- Organic Synthesis Group, Department of Chemistry and the Centre for Nanotechnology and Advanced Biomaterials, School of Chemical and Biotechnology, SASTRA University, Thanjavur 613401, Tamil Nadu, India
| | - Kumarasamy Muthusamy
- Organic Synthesis Group, Department of Chemistry and the Centre for Nanotechnology and Advanced Biomaterials, School of Chemical and Biotechnology, SASTRA University, Thanjavur 613401, Tamil Nadu, India
| | - Y Siva Prasad
- Organic Synthesis Group, Department of Chemistry and the Centre for Nanotechnology and Advanced Biomaterials, School of Chemical and Biotechnology, SASTRA University, Thanjavur 613401, Tamil Nadu, India
| | - Praveen Kumar Vemula
- Technologies for the Advancement of Science, Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences, UAS-GKVK Post, Bellary Road, Bangalore 560065, India
| | - Subbiah Nagarajan
- Organic Synthesis Group, Department of Chemistry and the Centre for Nanotechnology and Advanced Biomaterials, School of Chemical and Biotechnology, SASTRA University, Thanjavur 613401, Tamil Nadu, India.
| |
Collapse
|
15
|
Combination of hepatocyte specific delivery and transformation dependent expression of shRNA inducing transcriptional gene silencing of c-Myc promoter in hepatocellular carcinoma cells. BMC Cancer 2014; 14:582. [PMID: 25108398 PMCID: PMC4153911 DOI: 10.1186/1471-2407-14-582] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 07/28/2014] [Indexed: 12/11/2022] Open
Abstract
Background A specific targeting modality for hepatocellular carcinoma (HCC) could ideally encompass a liver cell specific delivery system of a transcriptional unit that is active only in neoplastic cells. Sendai virosomes, derived from Sendai viral envelopes, home to hepatocytes based on the liver specific expression of asialoglycoprotein receptors (ASGPRs) which are recognized by the Sendai virosomal fusion (F) proteins. As reported earlier by us and other groups, transcriptional gene silencing (TGS) does not require continuous presence of the effector siRNA/shRNA molecule and is heritable, involving epigenetic modifications, leading to long term transcriptional repression. This could be advantageous over conventional gene therapy approaches, since continuous c-Myc inactivation is required to suppress hepatocarcinoma cells. Methods Exploiting such virosomal delivery, the alpha-fetoprotein (AFP) promoter, in combination with various tumour specific enhancers, was used to drive the expression of shRNA directed against ME1a1 binding site of the proto-oncogene c-Myc P2 promoter, in order to induce TGS in neoplastic liver cells. Results The dual specificity achieved by the Sendai virosomal delivery system and the promoter/enhancer guided expression ensured that the shRNA inducing TGS was active only in liver cells that had undergone malignant transformation. Our results indicate that such a bimodal therapeutic system induced specific activation of apoptosis in hepatocarcinoma cells due to heterochromatization and increased DNA methylation of the CpG islands around the target loci. Conclusions The Sendai virosomal delivery system, combined with AFP promoter/enhancer expression machinery, could serve as a generalized mechanism for the expression of genes deleterious to transformed hepatocarcinoma cells. In this system, the epigenetic suppression of c-Myc could have an added advantage for inducing cell death in the targeted cells. Electronic supplementary material The online version of this article (doi:10.1186/1471-2407-14-582) contains supplementary material, which is available to authorized users.
Collapse
|
16
|
Schmidt S, Berens C, Klotzsche M. A novel TetR-regulating peptide turns off rtTA-mediated activation of gene expression. PLoS One 2014; 9:e96546. [PMID: 24810590 PMCID: PMC4014509 DOI: 10.1371/journal.pone.0096546] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 04/09/2014] [Indexed: 11/18/2022] Open
Abstract
Conditional regulation of gene expression is a powerful and indispensable method for analyzing gene function. The “Tet-On” system is a tool widely used for that purpose. Here, the transregulator rtTA mediates expression of a gene of interest after addition of the small molecule effector doxycycline. Although very effective in rapidly turning on gene expression, the system is hampered by the long half-life of doxycycline which makes shutting down gene expression rapidly very difficult to achieve. We isolated an rtTA-binding peptide by in vivo selection that acts as a doxycycline antagonist and leads to rapid and efficient shut down of rtTA-mediated reporter gene expression in a human cell line. This peptide represents the basis for novel effector molecules which complement the “Tet-system” by enabling the investigator to rapidly turn gene expression not just on at will, but now also off.
Collapse
Affiliation(s)
- Sebastian Schmidt
- Lehrstuhl für Mikrobiologie, Department Biologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Berens
- Lehrstuhl für Mikrobiologie, Department Biologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Marcus Klotzsche
- Lehrstuhl für Mikrobiologie, Department Biologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- * E-mail:
| |
Collapse
|
17
|
Niesner B, Maheshri N. Using the cre-lox system to randomize target gene expression states and generate diverse phenotypes. Biotechnol Bioeng 2013; 110:2677-86. [DOI: 10.1002/bit.24952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 04/03/2013] [Accepted: 04/22/2013] [Indexed: 11/06/2022]
Affiliation(s)
- Bradley Niesner
- Department of Chemical Engineering; Massachusetts Institute of Technology; Cambridge; Massachusetts; 02139
| | - Narendra Maheshri
- Department of Chemical Engineering; Massachusetts Institute of Technology; Cambridge; Massachusetts; 02139
| |
Collapse
|
18
|
Nevozhay D, Zal T, Balázsi G. Transferring a synthetic gene circuit from yeast to mammalian cells. Nat Commun 2013; 4:1451. [PMID: 23385595 PMCID: PMC3573884 DOI: 10.1038/ncomms2471] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 01/10/2013] [Indexed: 01/22/2023] Open
Abstract
The emerging field of synthetic biology builds gene circuits for scientific, industrial, and therapeutic needs. Adaptability of synthetic gene circuits across different organisms could enable a synthetic biology pipeline, where circuits are designed in silico, characterized in microbes and reimplemented in mammalian settings for practical usage. However, the processes affecting gene circuit adaptability have not been systematically investigated. Here we construct a mammalian version of a negative feedback-based “linearizer” gene circuit previously developed in yeast. The first naïve mammalian prototype was non-functional, but a computational model suggested that we could recover function by improving gene expression and protein localization. After rationally developing and combining new parts as the model suggested, we regained function and could tune target gene expression in human cells linearly and precisely as in yeast. The steps we have taken should be generally relevant for transferring any gene circuit from yeast into mammalian cells.
Collapse
Affiliation(s)
- Dmitry Nevozhay
- Department of Systems Biology-Unit 950, The University of Texas MD Anderson Cancer Center, 7435 Fannin Street, Houston, Texas 77054, USA
| | | | | |
Collapse
|
19
|
Jing Z, Gangalum RK, Lee JZ, Mock D, Bhat SP. Cell-type-dependent access of HSF1 and HSF4 to αB-crystallin promoter during heat shock. Cell Stress Chaperones 2013; 18:377-87. [PMID: 23264262 PMCID: PMC3631099 DOI: 10.1007/s12192-012-0386-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 10/18/2012] [Accepted: 11/13/2012] [Indexed: 10/27/2022] Open
Abstract
Epithelial cells and fibroblasts both express heat shock transcription factors, HSF1 and HSF4, yet they respond to heat shock differentially. For example, while HSP70 is induced in both cell types, the small heat shock protein, αB-crystallin gene (CRYAB) that contains a canonical heat shock promoter, is only induced in fibroblasts. A canonical heat shock promoter contains three or more inverted repeats of the pentanucleotide 5'-nGAAn-3' that make the heat shock element. It is known that, in vitro, promoter architecture (the order and spacing of these repeats) impacts the interaction of various heat shock transcription factors (HSFs) with the heat shock promoter, but in vivo relevance of these binding preferences so far as the expression is concerned is poorly understood. In this report, we first establish cell-type-dependent differential expression of CRYAB in four established cell lines and then working with adult human retinal pigment epithelial cells and NIH3T3 fibroblasts and employing chromatin immunoprecipitation, attempt to relate expression to promoter occupancy by HSF1 and HSF4. We show that HSF4 occupies only CRYAB and not HSP70 promoter in epithelial cells, while HSF1 occupies only HSP70 promoter in both cell types, and cryab promoter, only in heat shocked fibroblasts; HSF4, on the other hand, is never seen on these two promoters in NIH3T3 fibroblasts. This comparative analysis with CRYAB and HSP70 demonstrates that differential heat shock response is controlled by cell-type-dependent access of HSFs (HSF1 and HSF4) to specific promoters, independent of the promoter architecture.
Collapse
Affiliation(s)
- Zhe Jing
- />Jules Stein Eye Institute, David Geffen School of Medicine at UCLA, Brain Research Institute and Molecular Biology Institute, University of California, Los Angeles, CA 90095 USA
| | - Rajendra K. Gangalum
- />Jules Stein Eye Institute, David Geffen School of Medicine at UCLA, Brain Research Institute and Molecular Biology Institute, University of California, Los Angeles, CA 90095 USA
| | - Josh Z. Lee
- />Jules Stein Eye Institute, David Geffen School of Medicine at UCLA, Brain Research Institute and Molecular Biology Institute, University of California, Los Angeles, CA 90095 USA
| | - Dennis Mock
- />Jules Stein Eye Institute, David Geffen School of Medicine at UCLA, Brain Research Institute and Molecular Biology Institute, University of California, Los Angeles, CA 90095 USA
| | - Suraj P. Bhat
- />Jules Stein Eye Institute, David Geffen School of Medicine at UCLA, Brain Research Institute and Molecular Biology Institute, University of California, Los Angeles, CA 90095 USA
- />Jules Stein Eye Institute, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095 USA
| |
Collapse
|
20
|
Genetic Engineering of Mice. Mol Pharmacol 2012. [DOI: 10.1002/9781118451908.ch10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
21
|
Tai K, Quintino L, Isaksson C, Gussing F, Lundberg C. Destabilizing domains mediate reversible transgene expression in the brain. PLoS One 2012; 7:e46269. [PMID: 23029456 PMCID: PMC3460874 DOI: 10.1371/journal.pone.0046269] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 08/28/2012] [Indexed: 11/18/2022] Open
Abstract
Regulating transgene expression in vivo by delivering oral drugs has been a long-time goal for the gene therapy field. A novel gene regulating system based on targeted proteasomal degradation has been recently developed. The system is based on a destabilizing domain (DD) of the Escherichia coli dihydrofolate reductase (DHFR) that directs fused proteins to proteasomal destruction. Creating YFP proteins fused to destabilizing domains enabled TMP based induction of YFP expression in the brain, whereas omission of TMP resulted in loss of YFP expression. Moreover, induction of YFP expression was dose dependent and at higher TMP dosages, induced YFP reached levels comparable to expression of unregulated transgene., Transgene expression could be reversibly regulated using the DD system. Importantly, no adverse effects of TMP treatment or expression of DD-fusion proteins in the brain were observed. To show proof of concept that destabilizing domains derived from DHFR could be used with a biologically active molecule, DD were fused to GDNF, which is a potent neurotrophic factor of dopamine neurons. N-terminal placement of the DD resulted in TMP-regulated release of biologically active GDNF. Our findings suggest that TMP-regulated destabilizing domains can afford transgene regulation in the brain. The fact that GDNF could be regulated is very promising for developing future gene therapies (e.g. for Parkinson's disease) and should be further investigated.
Collapse
Affiliation(s)
| | | | | | | | - Cecilia Lundberg
- CNS Gene Therapy Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
22
|
Liu L, Abdel Motaal B, Schmidt-Supprian M, Pohl NLB. Multigram synthesis of isobutyl-β-C-galactoside as a substitute of isopropylthiogalactoside for exogenous gene induction in mammalian cells. J Org Chem 2012; 77:1539-46. [PMID: 22283618 DOI: 10.1021/jo2024569] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Herein we report that isobutyl-β-C-galactoside (IBCG) is also a promising inducer of gene expression in mammalian cells and report a new synthetic route to the compound that should make obtaining the multigram quantities of material required for animal studies more feasible. A convenient synthesis of IBCG, an inducer of genes controlled by the lac operon system in bacterial cells, was achieved in 5 steps from galactose in 81% overall yield without any chromatographic separation steps. An optimized microwave-assisted reaction at high concentration was key to making the C-glycosidic linkage. A Wittig reaction on a per-O-silylated rather than per-O-acetylated or -benzylated substrate proved most effective in installing the final carbon atom.
Collapse
Affiliation(s)
- Lin Liu
- Department of Chemistry, Plant Sciences Institute, Hach Hall, Iowa State University, Ames, Iowa 50011-3111, USA
| | | | | | | |
Collapse
|
23
|
Mehta V, Abi Nader K, Waddington S, David AL. Organ targeted prenatal gene therapy--how far are we? Prenat Diagn 2011; 31:720-34. [PMID: 21618255 DOI: 10.1002/pd.2787] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 04/14/2011] [Accepted: 04/17/2011] [Indexed: 12/14/2022]
Abstract
Prenatal gene therapy aims to deliver genes to cells and tissues early in prenatal life, allowing correction of a genetic defect, before long-term tissue damage has occurred. In contrast to postnatal gene therapy, prenatal application can target genes to a large population of dividing stem cells, and the smaller fetal size allows a higher vector-to-target cell ratio to be achieved. Early-gestation delivery may allow the development of immune tolerance to the transgenic protein which would facilitate postnatal repeat vector administration if needed. Targeting particular organs will depend on manipulating the vector to achieve selective tropism and on choosing the most appropriate gestational age and injection method for fetal delivery. Intra-amniotic injection reaches the skin, and other organs that are bathed in the fluid however since gene transfer to the lung and gut is usually poor more direct injection methods will be needed. Delivery to the liver and blood can be achieved by systemic delivery via the umbilical vein or peritoneal cavity. Gene transfer to the central nervous system in the fetus is difficult but newer vectors are available that transduce neuronal tissue even after systemic delivery.
Collapse
Affiliation(s)
- Vedanta Mehta
- Prenatal Cell and Gene Therapy Group, Institute for Women's Health, University College London, London, UK
| | | | | | | |
Collapse
|
24
|
König N, Åkesson E, Telorack M, Vasylovska S, Ngamjariyawat A, Sundström E, Oster A, Trolle C, Berens C, Aldskogius H, Seiger Å, Kozlova EN. Forced Runx1 expression in human neural stem/progenitor cells transplanted to the rat dorsal root ganglion cavity results in extensive axonal growth specifically from spinal cord-derived neurospheres. Stem Cells Dev 2011; 20:1847-57. [PMID: 21322790 DOI: 10.1089/scd.2010.0555] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cell replacement therapy holds great promise for treating a wide range of human disorders. However, ensuring the predictable differentiation of transplanted stem cells, eliminating their risk of tumor formation, and generating fully functional cells after transplantation remain major challenges in regenerative medicine. Here, we explore the potential of human neural stem/progenitor cells isolated from the embryonic forebrain (hfNSPCs) or the spinal cord (hscNSPCs) to differentiate to projection neurons when transplanted into the dorsal root ganglion cavity of adult recipient rats. To stimulate axonal growth, we transfected hfNSPC- and hscNSPC-derived neurospheres, prior to their transplantation, with a Tet-Off Runx1-overexpressing plasmid to maintain Runx1 expression in vivo after transplantation. Although pronounced cell differentiation was found in the Runx1-expressing transplants from both cell sources, we observed extensive, long-distance growth of axons exclusively from hscNSPC-derived transplants. These axons ultimately reached the dorsal root transitional zone, the boundary separating peripheral and central nervous systems. Our data show that hscNSPCs have the potential to differentiate to projection neurons with long-distance axonal outgrowth and that Runx1 overexpression is a useful approach to induce such outgrowth in specific sources of NSPCs.
Collapse
Affiliation(s)
- Niclas König
- Department of Neuroscience, Neuroanatomy, Uppsala University Biomedical Center, Uppsala, Sweden
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Liu ML, Oh JS, An SS, Pennant WA, Kim HJ, Gwak SJ, Yoon DH, Kim KN, Lee M, Ha Y. Controlled nonviral gene delivery and expression using stable neural stem cell line transfected with a hypoxia-inducible gene expression system. J Gene Med 2010; 12:990-1001. [DOI: 10.1002/jgm.1527] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
26
|
Touchard E, Kowalczuk L, Bloquel C, Naud MC, Bigey P, Behar-Cohen F. The ciliary smooth muscle electrotransfer: basic principles and potential for sustained intraocular production of therapeutic proteins. J Gene Med 2010; 12:904-19. [DOI: 10.1002/jgm.1517] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
27
|
Abstract
Gene doping, the abuse of gene therapy for illicit athletic enhancement, is perceived as a coming threat and is a prime concern to the anti-doping community. This doping technique represents a significant ethical challenge and there are concerns regarding its safety for athletes. This article presents the basics of gene doping, potential strategies for its detection and the role of promising new technologies in aiding detection efforts. These include the use of lab-on-a-chip techniques as well as nanoparticles to enhance the performance of current analytical methods and to develop new doping detection strategies.
Collapse
Affiliation(s)
- Mai M H Mansour
- Department of Chemistry and YJ-Science and Technology Research Center, The American University in Cairo, Cairo, Egypt
| | | |
Collapse
|
28
|
Kim YK, Kwon JT, Choi JY, Jiang HL, Arote R, Jere D, Je YH, Cho MH, Cho CS. Suppression of tumor growth in xenograft model mice by programmed cell death 4 gene delivery using folate-PEG-baculovirus. Cancer Gene Ther 2010; 17:751-60. [PMID: 20539318 DOI: 10.1038/cgt.2010.28] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Cancer gene therapy using tumor suppressor genes is considered to be an attractive approach for arresting cell growth and inducing apoptosis. Programmed cell death 4 (Pdcd4) is a tumor suppressor gene, which prevents tumorigenesis and tumor progression. To address the issue of whether expression of PDCD4 protein induces apoptosis in cancerous cells, the Pdcd4 gene was delivered using folate-PEG-baculovirus. Folate-PEG-baculovirus containing Pdcd4 gene (F-P-Bac-Pdcd4) was constructed by attachment of F-PEG to the baculovirus surface using chemical modification. The F-P-Bac-Pdcd4 showed enhanced transduction efficiency, efficiently expressed PDCD4 protein, and induced apoptosis in human epidermal carcinoma (KB) cells as compared with an unmodified baculovirus. In a tumor xenograft study, injection of F-P-Bac-Pdcd4 into tumors established from the KB cell line by subcutaneous implantation significantly suppressed tumor growth and induced apoptosis. Thus, this study shows a new baculovirus-mediated tumor suppressor gene delivery system for cancer therapy.
Collapse
Affiliation(s)
- Y-K Kim
- Department of Agricultural Biotechnology, Seoul National University, Seoul, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Bioinformatics in new generation flavivirus vaccines. J Biomed Biotechnol 2010; 2010:864029. [PMID: 20467477 PMCID: PMC2867002 DOI: 10.1155/2010/864029] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Revised: 12/21/2009] [Accepted: 03/02/2010] [Indexed: 12/22/2022] Open
Abstract
Flavivirus infections are the most prevalent arthropod-borne infections world wide, often causing severe disease especially among children, the elderly, and the immunocompromised. In the absence of effective antiviral treatment, prevention through vaccination would greatly reduce morbidity and mortality associated with flavivirus infections. Despite the success of the empirically developed vaccines against yellow fever virus, Japanese encephalitis virus and tick-borne encephalitis virus, there is an increasing need for a more rational design and development of safe and effective vaccines. Several bioinformatic tools are available to support such rational vaccine design. In doing so, several parameters have to be taken into account, such as safety for the target population, overall immunogenicity of the candidate vaccine, and efficacy and longevity of the immune responses triggered. Examples of how bio-informatics is applied to assist in the rational design and improvements of vaccines, particularly flavivirus vaccines, are presented and discussed.
Collapse
|
30
|
Yoshitake K, Aoyagi H, Fujiwara H. Creation of a novel telomere-cutting endonuclease based on the EN domain of telomere-specific non-long terminal repeat retrotransposon, TRAS1. Mob DNA 2010; 1:13. [PMID: 20359340 PMCID: PMC2868851 DOI: 10.1186/1759-8753-1-13] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2009] [Accepted: 04/01/2010] [Indexed: 12/25/2022] Open
Abstract
Background The ends of chromosomes, termed telomeres consist of repetitive DNA. The telomeric sequences shorten with cell division and, when telomeres are critically abbreviated, cells stop proliferating. However, in cancer cells, by the expression of telomerase which elongates telomeres, the cells can continue proliferating. Many approaches for telomere shortening have been pursued in the past, but to our knowledge, cutting telomeres in vivo has not so far been demonstrated. In addition, there is lack of information on the cellular effects of telomere shortening in human cells. Results Here, we created novel chimeric endonucleases to cut telomeres by fusing the endonuclease domain (TRAS1EN) of the silkworm's telomere specific non-long terminal repeat retrotransposon TRAS1 to the human telomere-binding protein, TRF1. An in vitro assay demonstrated that the TRAS1EN-TRF1 chimeric endonucleases (T-EN and EN-T) cut the human (TTAGGG)n repeats specifically. The concentration of TRAS1EN-TRF1 chimeric endonucleases necessary for the cleavage of (TTAGGG)n repeats was about 40-fold lower than that of TRAS1EN alone. When TRAS1EN-TRF1 endonucleases were introduced into human U2OS cancer cells using adenovirus vectors, the enzymes localized at telomeres of nuclei, cleaved and shortened the telomeric DNA by double-strand breaks. When human U2OS and HFL-1 fibroblast cells were infected with EN-T recombinant adenovirus, their cellular proliferation was suppressed for about 2 weeks after infection. In contrast, the TRAS1EN mutant (H258A) chimeric endonuclease fused with TRF1 (ENmut-T) did not show the suppression effect. The EN-T recombinant adenovirus induced telomere shortening in U2OS cells, activated the p53-dependent pathway and caused the senescence associated cellular responses, while the ENmut-T construct did not show such effects. Conclusions A novel TRAS1EN-TRF1 chimeric endonuclease (EN-T) cuts the human telomeric repeats (TTAGGG)n specifically in vitro and in vivo. Thus, the chimeric endonuclease which is expressed from an adenoviral vector can suppress cell proliferation of cancer cells.
Collapse
Affiliation(s)
- Kazutoshi Yoshitake
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Bioscience Bldg 501, Kashiwa, 277-8562, Japan.
| | | | | |
Collapse
|
31
|
Ziauddin MF, Guo ZS, O'Malley ME, Austin F, Popovic PJ, Kavanagh MA, Li J, Sathaiah M, Thirunavukarasu P, Fang B, Lee YJ, Bartlett DL. TRAIL gene-armed oncolytic poxvirus and oxaliplatin can work synergistically against colorectal cancer. Gene Ther 2010; 17:550-9. [PMID: 20182517 PMCID: PMC3250063 DOI: 10.1038/gt.2010.5] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
We have explored a unique combination therapy for metastatic colorectal cancer. This strategy combines a potent and new oncolytic poxvirus expressing a membrane-bound tumor necrosis factor-related apoptosis-inducing ligand (TRAIL or TNFSF10) and oxaliplatin (Ox) chemotherapy. We hypothesized that TRAIL expression would increase the efficacy of the oncolytic poxvirus, and that the therapeutic efficacy would be further enhanced by combination with chemotherapy. The cytotoxicity to cancer cells by Ox, oncolytic vaccinia virus (VV) and trail gene-armed VV alone or in combination was tested in vitro. The trail gene armed oncolytic VV-expressed high levels of TRAIL in infected cancer cells and had greater potency as a cytotoxic agent compared with the parent VV. Ox alone exerted concentration-dependent cytotoxicity. In vitro, the combination of the two agents applied at suboptimal concentrations for individual therapy displayed synergy in inducing cancer cells into enhanced levels of apoptosis/necrosis. Western blot analyses were consistent with the notion that TRAIL induced cancer cell death mainly through apoptosis, whereas Ox and vJS6 induced cell death more through non-apoptotic death pathways. In two aggressive colorectal carcinomatosis models derived from human HCT116 and murine MC38 cells, the combination therapy displayed synergistic or additive antitumor activity and prolonged the survival of the tumor-bearing mice compared with either Ox chemotherapy or vvTRAIL-mediated oncolytic gene therapy alone. This combination strategy may provide a new avenue to treating peritoneal carcinomatosis and other types of metastases of colorectal cancer.
Collapse
Affiliation(s)
- M F Ziauddin
- Department of Surgery, University of Pittsburgh School of Medicine and University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Christensen M, Larsen LA, Kauppinen S, Schratt G. Recombinant Adeno-Associated Virus-Mediated microRNA Delivery into the Postnatal Mouse Brain Reveals a Role for miR-134 in Dendritogenesis in Vivo. Front Neural Circuits 2010; 3:16. [PMID: 20126250 PMCID: PMC2809579 DOI: 10.3389/neuro.04.016.2009] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Accepted: 10/07/2009] [Indexed: 12/21/2022] Open
Abstract
Recent studies using primary neuronal cultures have revealed important roles of the microRNA pathway in the regulation of neuronal development and morphology. For example, miR-134 is involved in dendritogenesis and spine development in hippocampal neurons by regulating local mRNA translation in dendrites. The in vivo roles of microRNAs in these processes are still uninvestigated, partly due to the lack of tools enabling stable in vivo delivery of microRNAs or microRNA inhibitors into neurons of the mammalian brain. Here we describe the construction and validation of a vector-based tool for stable delivery of microRNAs in vivo by use of recombinant adeno-associated virus (rAAV). rAAV-mediated overexpression of miR-134 in neurons of the postnatal mouse brain provided evidence for a negative role of miR-134 in dendritic arborization of cortical layer V pyramidal neurons in vivo, thereby confirming previous findings obtained with cultured neurons. Our system provides researchers with a unique tool to study the role of any candidate microRNA in vivo and can easily be adapted to microRNA loss-of-function studies. This platform should therefore greatly facilitate investigations on the role of microRNAs in synapse development, plasticity and behavior in vivo.
Collapse
Affiliation(s)
- Mette Christensen
- Interdisziplinäres Zentrum für Neurowissenschaften, SFB488 Junior Group, Institut für Neuroanatomie, Universitätsklinikum Heidelberg Heidelberg, Germany
| | | | | | | |
Collapse
|
33
|
Therapeutic Nucleic Acids. Gene Ther 2010. [DOI: 10.1007/978-88-470-1643-9_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
34
|
Connolly RJ, Lopez GA, Hoff AM, Jaroszeski MJ. Plasma facilitated delivery of DNA to skin. Biotechnol Bioeng 2009; 104:1034-40. [PMID: 19557830 DOI: 10.1002/bit.22451] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Non-viral delivery of cell-impermeant drugs and DNA in vivo has traditionally relied upon either chemical or physical stress applied directly to target tissues. Physical methods typically use contact between an applicator, or electrode, and the target tissue and may involve patient discomfort. To overcome contact-dependent limitations of such delivery methodologies, an atmospheric helium plasma source was developed to deposit plasma products onto localized treatment sites. Experiments performed in murine skin showed that samples injected with plasmid DNA encoding luciferase and treated with plasma demonstrated increased levels of expression relative to skin samples that received injections of DNA alone. Increased response relative to injection alone was observed when either positive or negative voltage was used to generate the helium plasma. Quantitative results over a 26-day follow-up period showed that luciferase levels as high as 19-fold greater than the levels obtained by DNA injection alone could be achieved. These findings indicate that plasmas may compete with other physical delivery methodologies when skin is the target tissue.
Collapse
Affiliation(s)
- Richard J Connolly
- Department of Chemical and Biomedical Engineering, University of South Florida, ENB 118, Tampa, Florida 33620, USA
| | | | | | | |
Collapse
|
35
|
Abstract
Synthetic biology aims to engineer novel cellular functions by assembling well-characterized molecular parts (i.e., nucleic acids and proteins) into biological "devices" that exhibit predictable behavior. Recently, efforts in eukaryotic synthetic biology have sprung from foundational work in bacteria. Designing synthetic circuits to operate reliably in the context of differentiating and morphologically complex cells presents unique challenges and opportunities for progress in the field. This review surveys recent advances in eukaryotic synthetic biology and describes how synthetic systems can be linked to natural cellular processes in order to manipulate cell behavior and to foster new discoveries in cell biology research.
Collapse
Affiliation(s)
- Karmella A Haynes
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA.
| | | |
Collapse
|
36
|
PiggyBac transposon-based inducible gene expression in vivo after somatic cell gene transfer. Mol Ther 2009; 17:2115-20. [PMID: 19809403 DOI: 10.1038/mt.2009.234] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Somatic cell gene transfer has permitted inducible gene expression in vivo through coinfection of multiple viruses. We hypothesized that the highly efficient plasmid-based piggyBac transposon system would enable long-term inducible gene expression in mice in vivo. We used a multiple-transposon delivery strategy to create a tetracycline-inducible expression system in vitro in human cells by delivering the two genes on separate transposons for inducible reporter gene expression along with a separate selectable transposon marker. Evaluation of stable cell lines revealed 100% of selected clones exhibited inducible expression via stable expression from three separate transposons simultaneously. We next tested and found that piggyBac-mediated gene transfer to liver or lung could achieve stable reporter gene expression in mice in vivo in either immunocompetent or immune deficient animals. A single injection of piggyBac transposons could achieve long-term inducible gene expression in the livers of mice in vivo, confirming our multiple-transposon strategy used in cultured cells. The plasmid-based piggyBac transposon system enables constitutive or inducible gene expression in vivo for potential therapeutic and biological applications without using viral vectors.
Collapse
|
37
|
Park YG. [Inhibition of hepatitis B virus replication by RNA interference]. THE KOREAN JOURNAL OF HEPATOLOGY 2009; 15:1-6. [PMID: 19346780 DOI: 10.3350/kjhep.2009.15.1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
|
38
|
Sousa F, Passarinha L, Queiroz J. Biomedical application of plasmid DNA in gene therapy: A new challenge for chromatography. Biotechnol Genet Eng Rev 2009. [DOI: 10.5661/bger-26-83] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|