1
|
Cai Q, Guo R, Chen D, Deng Z, Gao J. SynBioNanoDesign: pioneering targeted drug delivery with engineered nanomaterials. J Nanobiotechnology 2025; 23:178. [PMID: 40050980 PMCID: PMC11884119 DOI: 10.1186/s12951-025-03254-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 02/19/2025] [Indexed: 03/10/2025] Open
Abstract
Synthetic biology and nanotechnology fusion represent a transformative approach promoting fundamental and clinical biomedical science development. In SynBioNanoDesign, biological systems are reimagined as dynamic and programmable materials to yield engineered nanomaterials with emerging and specific functionalities. This review elucidates a comprehensive examination of synthetic biology's pivotal role in advancing engineered nanomaterials for targeted drug delivery systems. It begins with exploring the fundamental synergy between synthetic biology and nanotechnology, then highlights the current landscape of nanomaterials in targeted drug delivery applications. Subsequently, the review discusses the design of novel nanomaterials informed by biological principles, focusing on expounding the synthetic biology tools and the potential for developing advanced nanomaterials. Afterward, the research advances of innovative materials design through synthetic biology were systematically summarized, emphasizing the integration of genetic circuitry to program nanomaterial responses. Furthermore, the challenges, current weaknesses and opportunities, prospective directions, and ethical and societal implications of SynBioNanoDesign in drug delivery are elucidated. Finally, the review summarizes the transformative impact that synthetic biology may have on drug-delivery technologies in the future.
Collapse
Affiliation(s)
- Qian Cai
- State Key Lab of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, Fujian, China
| | - Rui Guo
- National and Local United Engineering Laboratory of Natural Biotoxin, College of Bee and Biomedical Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Dafu Chen
- National and Local United Engineering Laboratory of Natural Biotoxin, College of Bee and Biomedical Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Zixin Deng
- State Key Laboratory of Microbial Metabolism, Joint International Laboratory on Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Jiangtao Gao
- National and Local United Engineering Laboratory of Natural Biotoxin, College of Bee and Biomedical Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
| |
Collapse
|
2
|
Chang L, Duan W, Wang C, Zhang J. miRNA-126 Inhibits Osteogenic Differentiation of Rat Bone Marrow Mesenchymal Stem Cells (BMSCs). J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study was to determine whether microRNA (miRNA)-126 regulates osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). Rat BMSCs were extracted and stimulated for osteogenic differentiation. Functional experiments were conducted to assess miR-126’s impact
on BMSCs differentiation. Western blot and RT-qPCR determined miR-126 expression. ALP activity detection and alizarin red staining detection were also performed. After osteogenic differentiation of BMSCs, miR-126 expression was gradually decreased over time. Overexpression of miR-26 decreased
ALP activity, Notch signaling activity as well as declined Runx2 expression and calcium Salt nodules after treatment. Importantly, we found that Smad4 serves as a target of miR-126 while upregulation of the miRNA was accompanied with the decreased Smad4 protein expression without affecting
the Smad4 mRNA level. In conclusion, miR-126 restrains osteogenic differentiation through inhibition of SMAD4 signaling, providing a novel insight into the mechanism.
Collapse
Affiliation(s)
- Le Chang
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shaanxi, 710032, China
| | - Wei Duan
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shaanxi, 710032, China
| | - Chuang Wang
- Department of Orthopedics, The Ninth Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710054, China
| | - Jian Zhang
- Department of Orthopedics, The Ninth Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710054, China
| |
Collapse
|
3
|
Mianné J, Ahmed E, Bourguignon C, Fieldes M, Vachier I, Bourdin A, Assou S, De Vos J. Induced Pluripotent Stem Cells for Primary Ciliary Dyskinesia Modeling and Personalized Medicine. Am J Respir Cell Mol Biol 2019; 59:672-683. [PMID: 30230352 DOI: 10.1165/rcmb.2018-0213tr] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Primary ciliary dyskinesia (PCD) is a rare and heterogeneous genetic disorder that affects the structure and function of motile cilia. In the airway epithelium, impaired ciliary motion results in reduced or absent mucociliary clearance that leads to the appearance of chronic airway infection, sinusitis, and bronchiectasis. Currently, there is no effective treatment for PCD, and research is limited by the lack of convenient models to study this disease and investigate innovative therapies. Furthermore, the high heterogeneity of PCD genotypes is likely to hinder the development of a single therapy for all patients. The generation of patient-derived, induced pluripotent stem cells, and their differentiation into airway epithelium, as well as genome editing technologies, could represent major tools for in vitro PCD modeling and for developing personalized therapies. Here, we review PCD pathogenesis and then discuss how human induced pluripotent stem cells could be used to model this disease for the development of innovative, patient-specific biotherapies.
Collapse
Affiliation(s)
- Joffrey Mianné
- 1 Institute for Regenerative Medicine and Biotherapy, University of Montpellier, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire Montpellier, Montpellier, France
| | - Engi Ahmed
- 1 Institute for Regenerative Medicine and Biotherapy, University of Montpellier, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire Montpellier, Montpellier, France
| | - Chloé Bourguignon
- 1 Institute for Regenerative Medicine and Biotherapy, University of Montpellier, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire Montpellier, Montpellier, France
| | - Mathieu Fieldes
- 1 Institute for Regenerative Medicine and Biotherapy, University of Montpellier, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire Montpellier, Montpellier, France
| | - Isabelle Vachier
- 2 PhyMedExp, University of Montpellier, INSERM, Centre Hospitalier Universitaire Montpellier, Montpellier, France; and
| | - Arnaud Bourdin
- 2 PhyMedExp, University of Montpellier, INSERM, Centre Hospitalier Universitaire Montpellier, Montpellier, France; and
| | - Said Assou
- 1 Institute for Regenerative Medicine and Biotherapy, University of Montpellier, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire Montpellier, Montpellier, France
| | - John De Vos
- 1 Institute for Regenerative Medicine and Biotherapy, University of Montpellier, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire Montpellier, Montpellier, France.,3 Centre Hospitalier Universitaire Montpellier, Department of Cell and Tissue Engineering, Hospital Saint-Eloi, Montpellier, France
| |
Collapse
|
4
|
Öner A. Recent Advancements in Gene Therapy for Hereditary Retinal Dystrophies. Turk J Ophthalmol 2017; 47:338-343. [PMID: 29326851 PMCID: PMC5758769 DOI: 10.4274/tjo.41017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Accepted: 04/20/2017] [Indexed: 12/01/2022] Open
Abstract
Hereditary retinal dystrophies (HRDs) are degenerative diseases of the retina which have marked clinical and genetic heterogeneity. Common presentations among these disorders include night or colour blindness, tunnel vision, and subsequent progression to complete blindness. The known causative disease genes have a variety of developmental and functional roles, with mutations in more than 120 genes shown to be responsible for the phenotypes. In addition, mutations within the same gene have been shown to cause different disease phenotypes, even amongst affected individuals within the same family, highlighting further levels of complexity. The known disease genes encode proteins involved in retinal cellular structures, phototransduction, the visual cycle, and photoreceptor structure or gene regulation. Significant advancements have been made in understanding the genetic pathogenesis of ocular diseases, and gene replacement and gene silencing have been proposed as potentially efficacious therapies. Because of its favorable anatomical and immunological characteristics, the eye has been at the forefront of translational gene therapy. Recent improvements have been made in the safety and specificity of vector-based ocular gene transfer methods. Dozens of promising proofs of concept have been obtained in animal models of HRDs and some of them have been relayed to the clinic. The results from the first clinical trials for a congenital form of blindness have generated great interest and have demonstrated the safety and efficacy of intraocular administrations of viral vectors in humans. This review summarizes the clinical development of retinal gene therapy.
Collapse
Affiliation(s)
- Ayşe Öner
- Erciyes University Faculty of Medicine, Department of Ophthalmology, Kayseri, Turkey
| |
Collapse
|
5
|
Targeting Visceral Fat by Intraperitoneal Delivery of Novel AAV Serotype Vector Restricting Off-Target Transduction in Liver. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 6:68-78. [PMID: 28702474 PMCID: PMC5491462 DOI: 10.1016/j.omtm.2017.06.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 06/14/2017] [Indexed: 12/12/2022]
Abstract
It is challenging to genetically manipulate fat in adults. We demonstrate that intraperitoneal (i.p.) injection of an engineered adeno-associated virus (AAV) serotype Rec2 leads to high transduction of multiple visceral fat depots at a dose of 1 to 2 orders lower than commonly used doses for systemic gene delivery. To target adipose tissue, we develop a single AAV vector harboring two expression cassettes: one using the CBA promoter to drive transgene expression and one using the liver-specific albumin promoter to drive a microRNA-targeting WPRE sequence that only exists in this AAV vector. This dual-cassette vector achieves highly selective transduction of visceral fat while severely restricting off-target transduction of liver. As proof of efficacy, i.p. administration of an adipose-targeting Rec2 vector harboring the leptin gene corrects leptin deficiency, obesity, and metabolic syndromes of ob/ob mice. This study provides a powerful tool to genetically manipulate fat for basic research and gene therapies of genetic and acquired diseases.
Collapse
|
6
|
Sharif W, Sharif Z. Leber's congenital amaurosis and the role of gene therapy in congenital retinal disorders. Int J Ophthalmol 2017; 10:480-484. [PMID: 28393043 DOI: 10.18240/ijo.2017.03.24] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 08/19/2016] [Indexed: 11/23/2022] Open
Abstract
Leber's congenital amaurosis (LCA) and recent gene therapy advancement for treating inherited retinopathies were extensive literature reviewed using MEDLINE, PubMed and EMBASE. Adeno-associated viral vectors were the most utilised vectors for ocular gene therapy. Cone photoreceptor cells might use an alternate pathway which was not reliant of the retinal pigment epithelium (RPE) derived retinoid isomerohydrolase (RPE65) to access the 11-cis retinal dehydechromophore. Research efforts dedicated on the progression of a gene-based therapy for the treatment of LCA2. Such gene therapy approaches were extremely successful in canine, porcine and rodent LCA2 models. The recombinant AAV2.hRPE65v2 adeno-associated vector contained the RPE65 cDNA and was replication deficient. Its in vitro injection in target cells induced RPE65 protein production. The gene therapy trials that were so far conducted for inherited retinopathies have generated promising results. Phase I clinical trials to cure LCA and choroideremia demonstrated that adeno-associated viral vectors containing RPE genes and photoreceptors respectively, could be successfully administered to inherited retinopathy patients. A phase III trial is presently ongoing and if successful, it will lead the way to additional gene therapy attempts to cure monogenic, inherited retinopathies.
Collapse
Affiliation(s)
- Walid Sharif
- Department of Ophthalmology Birmingham & Midland Eye Centre, City Hospital NHS Trust, Birmingham B18 7QH, UK
| | - Zuhair Sharif
- Institute of Ophthalmology, University College London 11-43 Bath St, London EC1V 9EL, UK
| |
Collapse
|
7
|
Cao H, Ouyang H, Ip W, Du K, Duan W, Avolio J, Wu J, Duan C, Yeger H, Bear CE, Gonska T, Hu J, Moraes TJ. Testing gene therapy vectors in human primary nasal epithelial cultures. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2015; 2:15034. [PMID: 26730394 PMCID: PMC4685663 DOI: 10.1038/mtm.2015.34] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 07/27/2015] [Accepted: 08/05/2015] [Indexed: 01/01/2023]
Abstract
Cystic fibrosis (CF) results from mutations in the CF transmembrane conductance regulator (CFTR) gene, which codes for a chloride/bicarbonate channel in the apical epithelial membranes. CFTR dysfunction results in a multisystem disease including the development of life limiting lung disease. The possibility of a cure for CF by replacing defective CFTR has led to different approaches for CF gene therapy; all of which ultimately have to be tested in preclinical model systems. Primary human nasal epithelial cultures (HNECs) derived from nasal turbinate brushing were used to test the efficiency of a helper-dependent adenoviral (HD-Ad) vector expressing CFTR. HD-Ad-CFTR transduction resulted in functional expression of CFTR at the apical membrane in nasal epithelial cells obtained from CF patients. These results suggest that HNECs can be used for preclinical testing of gene therapy vectors in CF.
Collapse
Affiliation(s)
- Huibi Cao
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children , Toronto, Onatrio, Canada
| | - Hong Ouyang
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children , Toronto, Onatrio, Canada
| | - Wan Ip
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children , Toronto, Onatrio, Canada
| | - Kai Du
- Programme in Molecular Structure & Function, Research Institute, Hospital for Sick Children , Toronto, Ontario, Canada
| | - Wenming Duan
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children , Toronto, Onatrio, Canada
| | - Julie Avolio
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children , Toronto, Onatrio, Canada
| | - Jing Wu
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children , Toronto, Onatrio, Canada
| | - Cathleen Duan
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children , Toronto, Onatrio, Canada
| | - Herman Yeger
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children , Toronto, Onatrio, Canada
| | - Christine E Bear
- Programme in Molecular Structure & Function, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Tanja Gonska
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children, Toronto, Onatrio, Canada; Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Jim Hu
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children, Toronto, Onatrio, Canada; Programme in Molecular Structure & Function, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Theo J Moraes
- Programme in Physiology & Experimental Medicine, Research Institute, Hospital for Sick Children, Toronto, Onatrio, Canada; Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
8
|
Draghici B, Ilies MA. Synthetic Nucleic Acid Delivery Systems: Present and Perspectives. J Med Chem 2015; 58:4091-130. [PMID: 25658858 DOI: 10.1021/jm500330k] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Bogdan Draghici
- Department
of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, 3307 North Broad Street, Philadelphia, Pennsylvania 19140, United States
| | - Marc A. Ilies
- Department
of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, 3307 North Broad Street, Philadelphia, Pennsylvania 19140, United States
- Temple Materials Institute, 1803 North Broad Street, Philadelphia, Pennsylvania 19122, United States
| |
Collapse
|
9
|
Abstract
Gene therapy has been considered as the most ideal medical intervention for genetic diseases because it is intended to target the cause of diseases instead of disease symptoms. Availability of techniques for identification of genetic mutations and for in vitro manipulation of genes makes it practical and attractive. After the initial hype in 1990s and later disappointments in clinical trials for more than a decade, light has finally come into the tunnel in recent years, especially in the field of eye gene therapy where it has taken big strides. Clinical trials in gene therapy for retinal degenerative diseases such as Leber's congenital amaurosis (LCA) and choroideremia demonstrated clear therapeutic efficacies without apparent side effects. Although these successful examples are still rare and sporadic in the field, they provide the proof of concept for harnessing the power of gene therapy to treat genetic diseases and to modernize our medication. In addition, those success stories illuminate the path for the development of gene therapy treating other genetic diseases. Because of the differences in target organs and cells, distinct barriers to gene delivery exist in gene therapy for each genetic disease. It is not feasible for authors to review the current development in the entire field. Thus, in this article, we will focus on what we can learn from the current success in gene therapy for retinal degenerative diseases to speed up the gene therapy development for lung diseases, such as cystic fibrosis.
Collapse
|
10
|
O'Neill SM, Hinkle C, Chen SJ, Sandhu A, Hovhannisyan R, Stephan S, Lagor WR, Ahima RS, Johnston JC, Reilly MP. Targeting adipose tissue via systemic gene therapy. Gene Ther 2014; 21:653-61. [PMID: 24830434 PMCID: PMC4342115 DOI: 10.1038/gt.2014.38] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 02/18/2014] [Accepted: 03/18/2014] [Indexed: 02/06/2023]
Abstract
Adipose tissue has a critical role in energy and metabolic homeostasis, but it is challenging to adapt techniques to modulate adipose function in vivo. Here we develop an in vivo, systemic method of gene transfer specifically targeting adipose tissue using adeno-associated virus (AAV) vectors. We constructed AAV vectors containing cytomegalovirus promoter-regulated reporter genes, intravenously injected adult mice with vectors using multiple AAV serotypes, and determined that AAV2/8 best targeted adipose tissue. Altering vectors to contain adiponectin promoter/enhancer elements and liver-specific microRNA-122 target sites restricted reporter gene expression to adipose tissue. As proof of efficacy, the leptin gene was incorporated into the adipose-targeted expression vector, package into AAV2/8 and administered intravenously to 9- to 10-week-old ob/ob mice. Phenotypic changes were measured over an 8-week period. Leptin mRNA and protein were expressed in adipose and leptin protein was secreted into plasma. Mice responded with reversal of weight gain, decreased hyperinsulinemia and improved glucose tolerance. AAV2/8-mediated systemic delivery of an adipose-targeted expression vector can replace a gene lacking in adipose tissue and correct a mouse model of human disease, demonstrating experimental application and therapeutic potential in disorders of adipose.
Collapse
Affiliation(s)
- Sean M. O'Neill
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christine Hinkle
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shu-Jen Chen
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arbansjit Sandhu
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ruben Hovhannisyan
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen Stephan
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - William R. Lagor
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rexford S. Ahima
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- The Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Julie C. Johnston
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Muredach P. Reilly
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Correspondence should be addressed to Muredach P. Reilly Cardiovascular Institute Translational Research Center 3400 Civic Center Blvd, Bldg 421 11th floor, Room 11-136 Philadelphia, PA 19104 Tel: (215) 573-1214 Fax: (215) 746-7415
| |
Collapse
|
11
|
Abstract
Virtually all currently used therapeutic agents are small molecules, largely because the development and delivery of small molecule drugs is relatively straightforward. Small molecules have serious limitations: drugs of this type can be fairly good enzyme inhibitors, receptor ligands, or allosteric modulators. However, most cellular functions are mediated by protein interactions with other proteins, and targeting protein-protein interactions by small molecules presents challenges that are unlikely to be overcome with these compounds as the only tools. Recent advances in gene delivery techniques and characterization of cell type-specific promoters open the prospect of using reengineered signaling-biased proteins as next-generation therapeutics. The first steps in targeted engineering of proteins with desired functional characteristics look very promising. As quintessential scaffolds that act strictly via interactions with other proteins in the cell, arrestins represent a perfect model for the development of these novel therapeutic agents with enormous potential: custom-designed signaling proteins will allow us to tell the cell what to do and when to do it in a way it cannot disobey.
Collapse
|
12
|
Retina-specific gene excision by targeted expression of Cre recombinase. Biochem Biophys Res Commun 2013; 441:777-81. [PMID: 24211578 DOI: 10.1016/j.bbrc.2013.10.139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 10/25/2013] [Indexed: 10/26/2022]
Abstract
The use of Cre recombinase for conditional targeting permits the controlled removal or activation of genes in specific tissues and at specific times of development. The Rho-Cre mice provide an improved tool for studying gene ablation in rod photoreceptor cells. To establish a robust expression of Rho-Cre transgenic mice that would be useful for the study of various protein functions in photoreceptor cells, a total 11,987 kb fragment (pNCHS4 Rho-NLS-cre) containing human rhodopsin promoter was cloned. The Rho-Cre plasmid was digested with EcoR1 and I Ceu-1, and the 9.316 kb fragment containing the hRho promoter and Cre recombinase gel was purified. To generate transgenic mice, the purified DNA fragment was injected into fertilized oocytes according to standard protocols. ROSA26R reported the steady expression of Rho-Cre especially in photoreceptor cells, allowing further excising proteins in rod photoreceptors across the retina. This Rho-Cre transgenic line should thus prove useful as a general deletor line for genetic analysis of diverse aspects of retinopathy.
Collapse
|
13
|
Yuan XW, Ge XQ, Sun XT, Ding YT. Intravenous administration of endothelial progenitor cells transfected with the TRAIL gene inhibits the growth of tumors derived from H22 cells in nude mice. Shijie Huaren Xiaohua Zazhi 2012; 20:2986-2991. [DOI: 10.11569/wcjd.v20.i31.2986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the influence of intravenous administration of endothelial progenitor cells (EPCs) transfected with the TRAIL gene on the growth of tumors derived from subcutaneously inoculated H22 cells in nude mice to provide a theoretical basis for the treatment of liver cancer.
METHODS: The TRAIL gene was amplified by PCR, cloned into the pcDNA3.1 vector, and transfected into EPCs. The expression of TRAIL protein was detected by Western blot. Mice were inoculated subcutaneously with H22 cells to induce tumor formation. Tumor-bearing mice were randomly divided into three groups and injected via the tail vein with EPCs transfected with the recombinant adenoviral vector carrying the TRAIL gene, the empty vector, and normal saline, respectively.
RESULTS: Restriction enzyme digestion and DNA sequencing analyses indicate that the recombinant plasmid was constructed successfully. TRAIL expression was detected in EPCs transfected with the recombinant adenoviral vector by Western blot. The rate of reduced tumor growth was 47.77% in mice administered with EPCs carrying the TRAIL gene. Tumor volume and weight in the experimental group (0.791 cm3 ± 0.119 cm3, 0.29 g ± 0.04 g) were significantly lower than those in the two control groups (all P < 0.05).
CONCLUSION: The recombinant plasmid carrying the TRAIL gene has been successfully constructed. Intravenous administration of endothelial progenitor cells transfected with the TRAIL gene inhibits the growth of tumors derived from H22 cells in nude mice.
Collapse
|
14
|
Abstract
Accumulation of all-trans-retinal (all-trans-RAL), reactive vitamin A aldehyde, is one of the key factors in initiating retinal photodamage. This photodamage is characterized by progressive retinal cell death evoked by light exposure in both an acute and chronic fashion. Photoactivated rhodopsin releases all-trans-RAL, which is subsequently transported by ATP-binding cassette transporter 4 and reduced to all-trans-retinol by all-trans-retinol dehydrogenases located in photoreceptor cells. Any interruptions in the clearing of all-trans-RAL in the photoreceptors can cause an accumulation of this reactive aldehyde and its toxic condensation products. This accumulation may result in the manifestation of retinal dystrophy including human retinal degenerative diseases such as Stargardt's disease and age-related macular degeneration. Herein, we discuss the mechanisms of all-trans-RAL clearance in photoreceptor cells by sequential enzymatic reactions, the visual (retinoid) cycle, and potential molecular pathways of retinal photodamage. We also review recent imaging technologies to monitor retinal health status as well as novel therapeutic strategies preventing all-trans-RAL-associated retinal photodamage.
Collapse
Affiliation(s)
- Tadao Maeda
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, USA
| | | | | |
Collapse
|
15
|
Saudou F. A "so cilia" network: cilia proteins start "social" networking. J Clin Invest 2012; 122:1198-202. [PMID: 22446182 DOI: 10.1172/jci62971] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cilia are unique cellular organelles found in nearly all cell types. In recent years, the importance of these organelles has been highlighted by the discovery that mutations in genes encoding proteins related to cilia biogenesis and function cause a class of complex syndromes termed ciliopathies. Emerging evidence suggests interactions among the various ciliopathy-associated proteins, but the precise mechanisms by which these interactions generate functional networks have remained elusive. In this issue of the JCI, Rachel and colleagues have now clearly linked two ciliopathy-associated proteins (CEP290 and MKKS). Surprisingly, the effects of a hypomorphic disease-causing Cep290 allele were rescued by loss of MKKS function, suggesting that it might be possible to treat some ciliopathies by fine-tuning interactions within the expanding ciliary network.
Collapse
Affiliation(s)
- Frédéric Saudou
- Institut Curie, CNRS UMR3306, and INSERM U1005, Orsay, France.
| |
Collapse
|