51
|
Selvaraj UM, Zuurbier KR, Whoolery CW, Plautz EJ, Chambliss KL, Kong X, Zhang S, Kim SH, Katzenellenbogen BS, Katzenellenbogen JA, Mineo C, Shaul PW, Stowe AM. Selective Nonnuclear Estrogen Receptor Activation Decreases Stroke Severity and Promotes Functional Recovery in Female Mice. Endocrinology 2018; 159:3848-3859. [PMID: 30256928 PMCID: PMC6203892 DOI: 10.1210/en.2018-00600] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/18/2018] [Indexed: 12/21/2022]
Abstract
Estrogens provide neuroprotection in animal models of stroke, but uterotrophic effects and cancer risk limit translation. Classic estrogen receptors (ERs) serve as transcription factors, whereas nonnuclear ERs govern numerous cell processes and exert beneficial cardiometabolic effects without uterine or breast cancer growth in mice. Here, we determined how nonnuclear ER stimulation with pathway-preferential estrogen (PaPE)-1 affects stroke outcome in mice. Ovariectomized female mice received vehicle, estradiol (E2), or PaPE-1 before and after transient middle cerebral artery occlusion (tMCAo). Lesion severity was assessed with MRI, and poststroke motor function was evaluated through 2 weeks after tMCAo. Circulating, spleen, and brain leukocyte subpopulations were quantified 3 days after tMCAo by flow cytometry, and neurogenesis and angiogenesis were evaluated histologically 2 weeks after tMCAo. Compared with vehicle, E2 and PaPE-1 reduced infarct volumes at 3 days after tMCAo, though only PaPE-1 reduced leukocyte infiltration into the ischemic brain. Unlike E2, PaPE-1 had no uterotrophic effect. Both interventions had negligible effect on long-term poststroke neuronal or vascular plasticity. All mice displayed a decline in motor performance at 2 days after tMCAo, and vehicle-treated mice did not improve thereafter. In contrast, E2 and PaPE-1 treatment afforded functional recovery at 6 days after tMCAo and beyond. Thus, the selective activation of nonnuclear ER by PaPE-1 decreased stroke severity and improved functional recovery in mice without undesirable uterotrophic effects. The beneficial effects of PaPE-1 are also associated with attenuated neuroinflammation in the brain. PaPE-1 and similar molecules may warrant consideration as efficacious ER modulators providing neuroprotection without detrimental effects on the uterus or cancer risk.
Collapse
Affiliation(s)
- Uma Maheswari Selvaraj
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kielen R Zuurbier
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Cody W Whoolery
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Erik J Plautz
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ken L Chambliss
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Xiangmei Kong
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Shanrong Zhang
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sung Hoon Kim
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Benita S Katzenellenbogen
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | | | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ann M Stowe
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Neurology, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
52
|
Florendo M, Figacz A, Srinageshwar B, Sharma A, Swanson D, Dunbar GL, Rossignol J. Use of Polyamidoamine Dendrimers in Brain Diseases. Molecules 2018; 23:molecules23092238. [PMID: 30177605 PMCID: PMC6225146 DOI: 10.3390/molecules23092238] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/24/2018] [Accepted: 08/28/2018] [Indexed: 12/18/2022] Open
Abstract
Polyamidoamine (PAMAM) dendrimers are one of the smallest and most precise nanomolecules available today, which have promising applications for the treatment of brain diseases. Each aspect of the dendrimer (core, size or generation, size of cavities, and surface functional groups) can be precisely modulated to yield a variety of nanocarriers for delivery of drugs and genes to brain cells in vitro or in vivo. Two of the most important criteria to consider when using PAMAM dendrimers for neuroscience applications is their safety profile and their potential to be prepared in a reproducible manner. Based on these criteria, features of PAMAM dendrimers are described to help the neuroscience researcher to judiciously choose the right type of dendrimer and the appropriate method for loading the drug to form a safe and effective delivery system to the brain.
Collapse
Affiliation(s)
- Maria Florendo
- College of Medicine, Central Michigan University, Mt. Pleasant, MI 48859, USA.
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI 48859, USA.
| | - Alexander Figacz
- College of Medicine, Central Michigan University, Mt. Pleasant, MI 48859, USA.
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI 48859, USA.
| | - Bhairavi Srinageshwar
- College of Medicine, Central Michigan University, Mt. Pleasant, MI 48859, USA.
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI 48859, USA.
- Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI 48859, USA.
| | - Ajit Sharma
- Department of Chemistry & Biochemistry, Central Michigan University, Mt. Pleasant, MI 48859, USA.
| | - Douglas Swanson
- Department of Chemistry & Biochemistry, Central Michigan University, Mt. Pleasant, MI 48859, USA.
| | - Gary L Dunbar
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI 48859, USA.
- Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI 48859, USA.
- Department of Psychology, Central Michigan University, Mt. Pleasant, MI 48859, USA.
- Field Neurosciences Institute, St. Mary's of Michigan, Saginaw, MI 48604, USA.
| | - Julien Rossignol
- College of Medicine, Central Michigan University, Mt. Pleasant, MI 48859, USA.
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI 48859, USA.
- Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI 48859, USA.
| |
Collapse
|
53
|
Cwetsch AW, Pinto B, Savardi A, Cancedda L. In vivo methods for acute modulation of gene expression in the central nervous system. Prog Neurobiol 2018; 168:69-85. [PMID: 29694844 PMCID: PMC6080705 DOI: 10.1016/j.pneurobio.2018.04.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 04/17/2018] [Accepted: 04/20/2018] [Indexed: 12/17/2022]
Abstract
Accurate and timely expression of specific genes guarantees the healthy development and function of the brain. Indeed, variations in the correct amount or timing of gene expression lead to improper development and/or pathological conditions. Almost forty years after the first successful gene transfection in in vitro cell cultures, it is currently possible to regulate gene expression in an area-specific manner at any step of central nervous system development and in adulthood in experimental animals in vivo, even overcoming the very poor accessibility of the brain. Here, we will review the diverse approaches for acute gene transfer in vivo, highlighting their advantages and disadvantages with respect to the efficiency and specificity of transfection as well as to brain accessibility. In particular, we will present well-established chemical, physical and virus-based approaches suitable for different animal models, pointing out their current and future possible applications in basic and translational research as well as in gene therapy.
Collapse
Affiliation(s)
- Andrzej W Cwetsch
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy; Università degli Studi di Genova, Via Balbi, 5, 16126 Genova, Italy
| | - Bruno Pinto
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy; Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126, Pisa, Italy
| | - Annalisa Savardi
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy; Università degli Studi di Genova, Via Balbi, 5, 16126 Genova, Italy
| | - Laura Cancedda
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy; DulbeccoTelethon Institute, Italy.
| |
Collapse
|
54
|
Kumari P, Rompicharla SVK, Muddineti OS, Ghosh B, Biswas S. Transferrin-anchored poly(lactide) based micelles to improve anticancer activity of curcumin in hepatic and cervical cancer cell monolayers and 3D spheroids. Int J Biol Macromol 2018; 116:1196-1213. [DOI: 10.1016/j.ijbiomac.2018.05.040] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/06/2018] [Accepted: 05/08/2018] [Indexed: 12/29/2022]
|
55
|
Abstract
Polyglutamine diseases are hereditary degenerative disorders of the nervous system that have remained, to this date, untreatable. Promisingly, investigation into their molecular etiology and the development of increasingly perfected tools have contributed to the design of novel strategies with therapeutic potential. Encouraging studies have explored gene therapy as a means to counteract cell demise and loss in this context. The current chapter addresses the two main focuses of research in the area: the characteristics of the systems used to deliver nucleic acids to cells and the molecular and cellular actions of the therapeutic agents. Vectors used in gene therapy have to satisfyingly reach the tissues and cell types of interest, while eliciting the lowest toxicity possible. Both viral and non-viral systems have been developed for the delivery of nucleic acids to the central nervous system, each with its respective advantages and shortcomings. Since each polyglutamine disease is caused by mutation of a single gene, many gene therapy strategies have tried to halt degeneration by silencing the corresponding protein products, usually recurring to RNA interference. The potential of small interfering RNAs, short hairpin RNAs and microRNAs has been investigated. Overexpression of protective genes has also been evaluated as a means of decreasing mutant protein toxicity and operate beneficial alterations. Recent gene editing tools promise yet other ways of interfering with the disease-causing genes, at the most upstream points possible. Results obtained in both cell and animal models encourage further delving into this type of therapeutic strategies and support the future use of gene therapy in the treatment of polyglutamine diseases.
Collapse
|
56
|
Rodriguez‐Otormin F, Duro‐Castano A, Conejos‐Sánchez I, Vicent MJ. Envisioning the future of polymer therapeutics for brain disorders. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2018; 11:e1532. [DOI: 10.1002/wnan.1532] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/24/2018] [Accepted: 05/09/2018] [Indexed: 01/09/2023]
Affiliation(s)
| | - Aroa Duro‐Castano
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Valencia Spain
| | | | - María J. Vicent
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Valencia Spain
| |
Collapse
|
57
|
Preferential and Increased Uptake of Hydroxyl-Terminated PAMAM Dendrimers by Activated Microglia in Rabbit Brain Mixed Glial Culture. Molecules 2018; 23:molecules23051025. [PMID: 29702566 PMCID: PMC6102539 DOI: 10.3390/molecules23051025] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/18/2018] [Accepted: 04/20/2018] [Indexed: 01/05/2023] Open
Abstract
Polyamidoamine (PAMAM) dendrimers are multifunctional nanoparticles with tunable physicochemical features, making them promising candidates for targeted drug delivery in the central nervous system (CNS). Systemically administered dendrimers have been shown to localize in activated glial cells, which mediate neuroinflammation in the CNS. These dendrimers delivered drugs specifically to activated microglia, producing significant neurological improvements in multiple brain injury models, including in a neonatal rabbit model of cerebral palsy. To gain further insight into the mechanism of dendrimer cell uptake, we utilized an in vitro model of primary glial cells isolated from newborn rabbits to assess the differences in hydroxyl-terminated generation 4 PAMAM dendrimer (D4-OH) uptake by activated and non-activated glial cells. We used fluorescently-labelled D4-OH (D-Cy5) as a tool for investigating the mechanism of dendrimer uptake. D4-OH PAMAM dendrimer uptake was determined by fluorescence quantification using confocal microscopy and flow cytometry. Our results indicate that although microglial cells in the mixed cell population demonstrate early uptake of dendrimers in this in vitro system, activated microglia take up more dendrimer compared to resting microglia. Astrocytes showed delayed and limited uptake. We also illustrated the differences in mechanism of uptake between resting and activated microglia using different pathway inhibitors. Both resting and activated microglia primarily employed endocytotic pathways, which are enhanced in activated microglial cells. Additionally, we demonstrated that hydroxyl terminated dendrimers are taken up by primary microglia using other mechanisms including pinocytosis, caveolae, and aquaporin channels for dendrimer uptake.
Collapse
|
58
|
He D, Lin H, Yu Y, Shi L, Tu J. Precisely Defined Polymers for Efficient Gene Delivery. Top Curr Chem (Cham) 2018; 376:2. [PMID: 29335799 DOI: 10.1007/s41061-017-0183-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/27/2017] [Indexed: 01/03/2023]
|
59
|
Rodrigues DB, Oliveira JM, Santos TC, Reis RL. Dendrimers: Breaking the paradigm of current musculoskeletal autoimmune therapies. J Tissue Eng Regen Med 2018; 12:e1796-e1812. [DOI: 10.1002/term.2597] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 09/01/2017] [Accepted: 10/09/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Daniel B. Rodrigues
- 3B's Research Group – Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineUniversity of Minho Avepark 4805‐017 Barco Guimarães Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Joaquim M. Oliveira
- 3B's Research Group – Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineUniversity of Minho Avepark 4805‐017 Barco Guimarães Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision MedicineHeadquarters at University of Minho Avepark 4805‐017 Barco Guimarães Portugal
| | - Tírcia C. Santos
- 3B's Research Group – Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineUniversity of Minho Avepark 4805‐017 Barco Guimarães Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Rui L. Reis
- 3B's Research Group – Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineUniversity of Minho Avepark 4805‐017 Barco Guimarães Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision MedicineHeadquarters at University of Minho Avepark 4805‐017 Barco Guimarães Portugal
| |
Collapse
|
60
|
Zhang Y, Sun T, Jiang C. Biomacromolecules as carriers in drug delivery and tissue engineering. Acta Pharm Sin B 2018; 8:34-50. [PMID: 29872621 PMCID: PMC5985630 DOI: 10.1016/j.apsb.2017.11.005] [Citation(s) in RCA: 268] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/05/2017] [Accepted: 10/07/2017] [Indexed: 12/14/2022] Open
Abstract
Natural biomacromolecules have attracted increased attention as carriers in biomedicine in recent years because of their inherent biochemical and biophysical properties including renewability, nontoxicity, biocompatibility, biodegradability, long blood circulation time and targeting ability. Recent advances in our understanding of the biological functions of natural-origin biomacromolecules and the progress in the study of biological drug carriers indicate that such carriers may have advantages over synthetic material-based carriers in terms of half-life, stability, safety and ease of manufacture. In this review, we give a brief introduction to the biochemical properties of the widely used biomacromolecule-based carriers such as albumin, lipoproteins and polysaccharides. Then examples from the clinic and in recent laboratory development are summarized. Finally the current challenges and future prospects of present biological carriers are discussed.
Collapse
Key Words
- ABD, albumin binding domain
- ACM, aclacinomycin
- ACS, absorbable collagen sponge
- ADH, adipic dihydrazide
- ART, artemisinin
- ASF, Antheraea mylitta silk fibroin
- ATRA, all-trans retinoic acid
- ATS, artesunate
- BCEC, brain capillary endothelial cells
- BMP-2, bone morphogenetic protein-2
- BSA, bovine serum albumin
- BSF, Bombyx mori silk fibroin
- Biomacromolecule
- CC-HAM, core-crosslinked polymeric micelle based hyaluronic acid
- CD, cyclodextrin
- CD-NPs, amphiphilic MMA–tBA β-CD star copolymers that are capable of forming nanoparticles
- CD-g-CS, chitosan grafted with β-cyclodextrin
- CD/BP, cyclodextrin–bisphosphonate complexes
- CIA, collagen-induced arthritis
- CM, collagen matrices
- CMD-ChNP, carboxylmethyl dextran chitosan nanoparticle
- DHA, dihydroartesunate
- DOXO-EMCH, (6-maleimidocaproyl)hydrazone derivative of doxorubicin
- DOX–TRF, doxorubincin–transferrin conjugate
- DTX-HPLGA, HA coated PLGA nanoparticulate docetaxel
- Drug delivery
- ECM, extracellular matrix
- EMT, epithelial mesenchymal transition
- EPR, enhanced permeability and retention
- FcRn, neonatal Fc receptor
- GAG, glycosaminoglycan
- GC-DOX, glycol–chitosan–doxorubicin conjugate
- GDNF, glial-derived neurotrophic factor
- GO, grapheme oxide
- GSH, glutathione
- Gd, gadolinium
- HA, hyaluronic acid
- HA-CA, catechol-modified hyaluronic acid
- HCF, heparin-conjugated fibrin
- HDL, high density lipoprotein
- HEK, human embryonic kidney
- HSA, human serum albumin
- IDL, intermediate density lipoprotein
- INF, interferon
- LDL, low density lipoprotein
- LDLR, low density lipoprotein receptor
- LDV, leucine–aspartic acid–valine
- LMWH, low molecular weight heparin
- MSA, mouse serum albumin
- MTX–HSA, methotrexate–albumin conjugate
- NIR, near-infrared
- NSCLC, non-small cell lung cancer
- OP-Gel-NS, oxidized pectin-gelatin-nanosliver
- PEC, polyelectrolyte
- PTX, paclitaxel
- Polysaccharide
- Protein
- RES, reticuloendothelial system
- RGD, Arg–Gly–Asp peptide
- SF, silk fibroin
- SF-CSNP, silk fibroin modified chitosan nanoparticle
- SFNP, silk fibroin nanoparticle
- SPARC, secreted protein acidic and rich in cysteine
- TRAIL, tumor-necrosis factor-related apoptosis-inducing ligand
- Tf, transferrin
- TfR, transferrin receptor
- Tissue engineering
- VEGF, vascular endothelial growth factor
- VLDL, very low density lipoprotein
- pDNA, plasmid DNA
- rHDL, recombinant HDL
- rhEGF-2/HA, recombinant human fibroblast growth factor type 2 in a hyaluronic acid carrier
Collapse
Affiliation(s)
| | | | - Chen Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 200032, China
| |
Collapse
|
61
|
Ayatollahi S, Salmasi Z, Hashemi M, Askarian S, Oskuee RK, Abnous K, Ramezani M. Aptamer-targeted delivery of Bcl-xL shRNA using alkyl modified PAMAM dendrimers into lung cancer cells. Int J Biochem Cell Biol 2017; 92:210-217. [PMID: 29031805 DOI: 10.1016/j.biocel.2017.10.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/07/2017] [Accepted: 10/10/2017] [Indexed: 11/25/2022]
Abstract
RNAi-based gene therapy has been recently considered as a promising approach against cancer. Targeted delivery of drug, gene or therapeutic RNAi-based systems to tumor cells is one of the important issues in order to reduce side effects on normal cells. Several strategies have been developed to improve the safety and selectivity of cancer treatments including antibodies, peptides and recently aptamers with various attractive characteristics including higher target specificity, affinity and reduced toxicity. Here we described a novel targeted delivery platform comprising modified PAMAM with 10-bromodecanoic acid (10C) and 10C-PEG for improvement of transfection efficiency, AS1411 aptamer for targeting nucleolin ligand on target cancer cells and shRNA plasmid for specific knockdown of Bcl-xL protein. Modified vector could significantly improve the transfection efficiency even after covalent or non-covalent aptamer binding compared to the non-targeted vector in A549 cells. The results of gene silencing and apoptosis assay indicated that our targeted shRNA delivery system could efficiently down-regulate the Bcl-xL expression up to 25% and induce 14% late apoptosis in target cancer cells with strong cell selectivity. This study proposed a novel targeted non-viral system for shRNA-mediated gene-silencing in cancer cells.
Collapse
Affiliation(s)
- Sara Ayatollahi
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Salmasi
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Hashemi
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeedeh Askarian
- Research Center of Advanced Technologies in Medicine, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Kazemi Oskuee
- Targeted drug delivery research center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
62
|
Lembo D, Donalisio M, Civra A, Argenziano M, Cavalli R. Nanomedicine formulations for the delivery of antiviral drugs: a promising solution for the treatment of viral infections. Expert Opin Drug Deliv 2017; 15:93-114. [DOI: 10.1080/17425247.2017.1360863] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- David Lembo
- Department of Clinical and Biological Sciences, University of Torino, S. Luigi Gonzaga Hospital, Torino, Italy
| | - Manuela Donalisio
- Department of Clinical and Biological Sciences, University of Torino, S. Luigi Gonzaga Hospital, Torino, Italy
| | - Andrea Civra
- Department of Clinical and Biological Sciences, University of Torino, S. Luigi Gonzaga Hospital, Torino, Italy
| | - Monica Argenziano
- Department of Drug Science and Technology, University of Torino, Turin, Italy
| | - Roberta Cavalli
- Department of Drug Science and Technology, University of Torino, Turin, Italy
| |
Collapse
|
63
|
Zeng Y, Kurokawa Y, Win-Shwe TT, Zeng Q, Hirano S, Zhang Z, Sone H. Effects of PAMAM dendrimers with various surface functional groups and multiple generations on cytotoxicity and neuronal differentiation using human neural progenitor cells. J Toxicol Sci 2017; 41:351-70. [PMID: 27193728 DOI: 10.2131/jts.41.351] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Polyamidoamine (PAMAM) dendrimers have potential for biological applications as delivery systems for genes, drugs, and imaging agents into the brain, but their developmental neurotoxicity remains unknown. We investigated the effects of PAMAM dendrimers with various surface functional groups and multiple generations on neuronal differentiation using human neural progenitor cells at an equal mass concentration. Only PAMAM dendrimers containing amine (NH2) surface groups at concentrations of 10 μg/mL significantly reduced cell viability and neuronal differentiation, compared with non-amine-terminated dendrimers. PAMAM-NH2 with generation (G)3, G4, G5 G6, and G7 significantly decreased cell viability and inhibited neuronal differentiation from a concentration of 5 μg/mL, but G0, G1, and G2 dendrimers did not have any effect at this concentration. Cytotoxicity indices of PAMAM-NH2 dendrimers at 10 μg/mL correlated well with the zeta potentials of the particles. Surface group density and particle number in unit volume is more important characteristic than particle size to influence cytotoxicity for positive changed dendrimers. PAMAM-50% C12 at 1 μg/mL altered the expression level of the oxidative stress-related genes, ROR1, CYP26A1, and TGFB1, which is a DNA damage response gene. Our results indicate that PAMAM dendrimer exposure may have a surface charge-dependent adverse effect on neuronal differentiation, and that the effect may be associated with oxidative stress and DNA damage during development of neural cells.
Collapse
Affiliation(s)
- Yang Zeng
- Center for Environmental Risk Research, National Institute for Environmental Studies
| | | | | | | | | | | | | |
Collapse
|
64
|
Li B, Li Q, Mo J, Dai H. Drug-Loaded Polymeric Nanoparticles for Cancer Stem Cell Targeting. Front Pharmacol 2017; 8:51. [PMID: 28261093 PMCID: PMC5306366 DOI: 10.3389/fphar.2017.00051] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 01/24/2017] [Indexed: 12/15/2022] Open
Abstract
Cancer stem cells (CSCs) have been reported to play critical roles in tumor initiation, propagation, and regeneration of cancer. Nano-size vehicles are employed to deliver drugs to target the CSCs for cancer therapy. Polymeric nanoparticles have been considered as the most efficient vehicles for drug delivery due to their excellent pharmacokinetic properties. The CSCs specific antibodies or ligands can be conjugated onto the surface or interior of nanoparticles to successfully target and finally eliminate CSCs. In this review, we focus on the approaches of polymeric nanoparticles design for loading drug, and their potential application for CSCs targeting in cancer therapy.
Collapse
Affiliation(s)
- Binbin Li
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of TechnologyWuhan, China
- Biomedical Materials and Engineering Research Center of Hubei ProvinceWuhan, China
| | - Qinghua Li
- Department of Neurology, Affiliated Hospital of Guilin Medical UniversityGuilin, China
| | - Jingxin Mo
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of EducationGuangzhou, China
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| | - Honglian Dai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of TechnologyWuhan, China
- Biomedical Materials and Engineering Research Center of Hubei ProvinceWuhan, China
| |
Collapse
|
65
|
Zhan H, Jagtiani T, Liang JF. A new targeted delivery approach by functionalizing drug nanocrystals through polydopamine coating. Eur J Pharm Biopharm 2017; 114:221-229. [PMID: 28161549 DOI: 10.1016/j.ejpb.2017.01.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 01/08/2017] [Accepted: 01/09/2017] [Indexed: 12/25/2022]
Abstract
Tumor target specificity via chemotherapy is widely considered to be very effective on tumor treatment. For an ideal chemotherapeutic agent like Camptothecin (CPT) (CPT is the abbreviation for Camptothecin), improved therapeutic efficacy and high selectivity are equally important. Inspired by adhesive proteins in mussels, here we developed a novel tumor targeting peptide XQ1 grafted CPT nanocrystals with polydopamine coating as a spacer. In this study, CPT nanocrystals were coated by polymerization of dopamine that was induced by plasma-activated water under an acidic environment, and then the tumor targeting peptide was grafted onto polydopamine (PDA) (PDA is the abbreviation for polydopamine) coated CPT nanocrystals through catechol chemistry. The PDA layer had negligible effects on drug crystallinity and structure but resulted in drug nanocrystals with excellent dispersion properties, improved dissolution rate and drug stability by preventing water hydrolysis. More importantly, tumor targeting peptide XQ1 facilitated a rapid cross-membrane translocation of drug nanocrystals via receptor-mediated endocytosis, leading to efficient intracellular drug delivery. Moreover, this novel drug formulation demonstrated more potent anti-cancer activity against tumor cells in comparison with free CPT and naked CPT nanocrystals and exhibited high selectivity, all of which are attributed to the tumor target specificity property and inherent pH-dependent drug release behavior.
Collapse
Affiliation(s)
- Honglei Zhan
- Department of Biomedical Engineering, Chemistry, and Biological Sciences, Charles V. Schaefer School of Engineering and Sciences, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Tina Jagtiani
- Department of Biomedical Engineering, Chemistry, and Biological Sciences, Charles V. Schaefer School of Engineering and Sciences, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Jun F Liang
- Department of Biomedical Engineering, Chemistry, and Biological Sciences, Charles V. Schaefer School of Engineering and Sciences, Stevens Institute of Technology, Hoboken, NJ 07030, USA.
| |
Collapse
|
66
|
Leda AR, Dygert L, Bertrand L, Toborek M. Mouse Microsurgery Infusion Technique for Targeted Substance Delivery into the CNS via the Internal Carotid Artery. J Vis Exp 2017. [PMID: 28190068 DOI: 10.3791/54804] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Animal models of central nervous system (CNS) diseases and, consequently, blood-brain barrier disruption diseases, require the delivery of exogenous substances into the brain. These exogenous substances may induce injurious impact or constitute therapeutic strategy. The most common delivery methods of exogenous substances into the brain are based on systemic deliveries, such as subcutaneous or intravenous routes. Although commonly used, these approaches have several limitations, including low delivery efficacy into the brain. In contrast, surgical methods that locally deliver substances into the CNS are more specific and prevent the uptake of the exogenous substances by other organs. Several surgical methods for CNS delivery are available; however, they tend to be very traumatic. Here, we describe a mouse infusion microsurgery technique, which effectively delivers substances into the brain via the internal carotid artery, with minimal trauma and no interference with normal CNS functionality.
Collapse
Affiliation(s)
- Ana R Leda
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami
| | - Levy Dygert
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami
| | - Luc Bertrand
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami; The Jerzy Kukuczka Academy of Physical Education;
| |
Collapse
|
67
|
Tabasum S, Noreen A, Kanwal A, Zuber M, Anjum MN, Zia KM. Glycoproteins functionalized natural and synthetic polymers for prospective biomedical applications: A review. Int J Biol Macromol 2017; 98:748-776. [PMID: 28111295 DOI: 10.1016/j.ijbiomac.2017.01.078] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 01/05/2017] [Accepted: 01/16/2017] [Indexed: 02/06/2023]
Abstract
Glycoproteins have multidimensional properties such as biodegradability, biocompatibility, non-toxicity, antimicrobial and adsorption properties; therefore, they have wide range of applications. They are blended with different polymers such as chitosan, carboxymethyl cellulose (CMC), polyvinyl pyrrolidone (PVP), polycaprolactone (PCL), heparin, polystyrene fluorescent nanoparticles (PS-NPs) and carboxyl pullulan (PC) to improve their properties like thermal stability, mechanical properties, resistance to pH, chemical stability and toughness. Considering the versatile charateristics of glycoprotein based polymers, this review sheds light on synthesis and characterization of blends and composites of glycoproteins, with natural and synthetic polymers and their potential applications in biomedical field such as drug delivery system, insulin delivery, antimicrobial wound dressing uses, targeting of cancer cells, development of anticancer vaccines, development of new biopolymers, glycoproteome research, food product and detection of dengue glycoproteins. All the technical scientific issues have been addressed; highlighting the recent advancement.
Collapse
Affiliation(s)
- Shazia Tabasum
- Institute of Chemistry, Government College University, Faisalabad 38030, Pakistan
| | - Aqdas Noreen
- Institute of Chemistry, Government College University, Faisalabad 38030, Pakistan
| | - Arooj Kanwal
- Institute of Chemistry, Government College University, Faisalabad 38030, Pakistan
| | - Mohammad Zuber
- Institute of Chemistry, Government College University, Faisalabad 38030, Pakistan
| | | | - Khalid Mahmood Zia
- Institute of Chemistry, Government College University, Faisalabad 38030, Pakistan.
| |
Collapse
|
68
|
Shi W, Cui X, Shi J, Chen J, Wang Y. Overcoming the blood–brain barrier for glioma-targeted therapy based on an interleukin-6 receptor-mediated micelle system. RSC Adv 2017. [DOI: 10.1039/c7ra03208k] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
An interleukin-6 receptor-mediated micelle-drug system was prepared for cascade-targeting chemotherapy of glioma, which exhibited high BBB-crossing and glioma-targeted efficiency.
Collapse
Affiliation(s)
- Wei Shi
- Department of Neurosurgery
- Affiliated Hospital of Nantong University
- Nantong 226001
- China
| | - Xuexue Cui
- Center for Advanced Low-dimension Materials
- Donghua University
- Shanghai 201620
- China
| | - Jinlong Shi
- Department of Neurosurgery
- Affiliated Hospital of Nantong University
- Nantong 226001
- China
| | - Jian Chen
- Department of Neurosurgery
- Affiliated Hospital of Nantong University
- Nantong 226001
- China
| | - Yi Wang
- Center for Advanced Low-dimension Materials
- Donghua University
- Shanghai 201620
- China
- Center of Analysis and Measurement
| |
Collapse
|
69
|
Tan JKY, Sellers DL, Pham B, Pun SH, Horner PJ. Non-Viral Nucleic Acid Delivery Strategies to the Central Nervous System. Front Mol Neurosci 2016; 9:108. [PMID: 27847462 PMCID: PMC5088201 DOI: 10.3389/fnmol.2016.00108] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 10/11/2016] [Indexed: 12/11/2022] Open
Abstract
With an increased prevalence and understanding of central nervous system (CNS) injuries and neurological disorders, nucleic acid therapies are gaining promise as a way to regenerate lost neurons or halt disease progression. While more viral vectors have been used clinically as tools for gene delivery, non-viral vectors are gaining interest due to lower safety concerns and the ability to deliver all types of nucleic acids. Nevertheless, there are still a number of barriers to nucleic acid delivery. In this focused review, we explore the in vivo challenges hindering non-viral nucleic acid delivery to the CNS and the strategies and vehicles used to overcome them. Advantages and disadvantages of different routes of administration including: systemic injection, cerebrospinal fluid injection, intraparenchymal injection and peripheral administration are discussed. Non-viral vehicles and treatment strategies that have overcome delivery barriers and demonstrated in vivo gene transfer to the CNS are presented. These approaches can be used as guidelines in developing synthetic gene delivery vectors for CNS applications and will ultimately bring non-viral vectors closer to clinical application.
Collapse
Affiliation(s)
- James-Kevin Y Tan
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington Seattle, WA, USA
| | - Drew L Sellers
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington Seattle, WA, USA
| | - Binhan Pham
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington Seattle, WA, USA
| | - Suzie H Pun
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington Seattle, WA, USA
| | - Philip J Horner
- Center for Neuroregenerative Medicine, Houston Methodist Research Institute Houston, TX, USA
| |
Collapse
|
70
|
Luong D, Kesharwani P, Deshmukh R, Mohd Amin MCI, Gupta U, Greish K, Iyer AK. PEGylated PAMAM dendrimers: Enhancing efficacy and mitigating toxicity for effective anticancer drug and gene delivery. Acta Biomater 2016; 43:14-29. [PMID: 27422195 DOI: 10.1016/j.actbio.2016.07.015] [Citation(s) in RCA: 259] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 06/27/2016] [Accepted: 07/11/2016] [Indexed: 11/17/2022]
Abstract
UNLABELLED Poly(amidoamine) dendrimers (PAMAM) are well-defined, highly branched, nanoscale macromolecules with numerous active amine groups on the surface. PAMAM dendrimer can enhance the solubility of hydrophobic drugs, and with numerous reactive groups on the surface PAMAM dendrimer can be engineered with various functional groups for specific targeting ability. However, in physiological conditions, these amine groups are toxic to cells and limit the application of PAMAM. In the recent years, polyethylene glycol (PEG) conjugation has been the most widely used approach to reduce the toxicity of the active group on dendrimer surface. PEG molecules are known to be inert, non-immunogenic, and non-antigenic with a significant water solubility. PEGylated PAMAM-mediated delivery could not only overcome the limitations of dendrimer such as drug leakage, immunogenicity, hemolytic toxicity, systemic cytotoxicity but they also have the ability to enhance the solubilization of hydrophobic drugs and facilitates the potential for DNA transfection, siRNA delivery and tumor targeting. This review focuses on the recent developments on the application and influence of PEGylation on various biopharmaceutical properties of PAMAM dendrimers. STATEMENT OF SIGNIFICANCE It is well established that dendrimers have demonstrated promising potentials for drug delivery. However, the inherent toxicity poses challenges for its clinical translation. In this regard, PEGylation has helped mitigate some of the toxicity concerns of dendrimers and have paved the way forward for testing its translational potentials. The review is a collection of articles demonstrating the utility of PEGylation of the most studied PAMAM dendrimers. To our knowledge, this is a first such attempt to draw reader's attention, specifically, towards PEGylated PAMAM dendrimers.
Collapse
Affiliation(s)
- Duy Luong
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, United States
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, United States
| | - Rahul Deshmukh
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, United States
| | - Mohd Cairul Iqbal Mohd Amin
- Centre for Drug Delivery Research, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz 50300, Kuala Lumpur, Malaysia
| | - Umesh Gupta
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Kishangarh, Ajmer 305801, India
| | - Khaled Greish
- Aljawhara Center for Molecular Medicine, Arabian Gulf University, Bahrain
| | - Arun K Iyer
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, United States; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University, School of Medicine, Detroit, MI 48201, United States.
| |
Collapse
|
71
|
Chambliss KL, Barrera J, Umetani M, Umetani J, Kim SH, Madak-Erdogan Z, Huang L, Katzenellenbogen BS, Katzenellenbogen JA, Mineo C, Shaul PW. Nonnuclear Estrogen Receptor Activation Improves Hepatic Steatosis in Female Mice. Endocrinology 2016; 157:3731-3741. [PMID: 27552247 PMCID: PMC5045504 DOI: 10.1210/en.2015-1629] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Estrogens have the potential to afford atheroprotection, to prevent excess adiposity and its metabolic complications including insulin resistance, and to lessen hepatic steatosis. Cellular responses to estrogens occur through gene regulation by nuclear estrogen receptors (ERs), and through signal initiation by plasma membrane-associated ER. Leveraging the potentially favorable cardiometabolic actions of estrogens has been challenging, because their reproductive tract and cancer-promoting effects adversely impact the risk to benefit ratio of the therapy. In previous works, we discovered that an estrogen dendrimer conjugate (EDC) comprised of ethinyl-estradiol (E2) molecules linked to a poly(amido)amine dendrimer selectively activates nonnuclear ER, and in mice, EDC does not invoke a uterotrophic response or support ER-positive breast cancer growth. In the present investigation, we employed EDC to determine how selective nonnuclear ER activation impacts atherosclerosis, adiposity, glucose homeostasis, and hepatic steatosis in female mice. In contrast to E2, EDC did not blunt atherosclerosis in hypercholesterolemic apoE-/- mice. Also in contrast to E2, EDC did not prevent the increase in adiposity caused by Western diet feeding in wild-type mice, and it did not affect Western diet-induced glucose intolerance. However, E2 and EDC had comparable favorable effect on diet-induced hepatic steatosis, and this was related to down-regulation of fatty acid and triglyceride synthesis genes in the liver. Predictably, only E2 caused a uterotrophic response. Thus, although nonnuclear ER activation does not prevent atherosclerosis or diet-induced obesity or glucose intolerance, it may provide a potential new strategy to combat hepatic steatosis without impacting the female reproductive tract or increasing cancer risk.
Collapse
Affiliation(s)
- Ken L Chambliss
- Center for Pulmonary and Vascular Biology (K.L.C., J.B., M.U., J.U., L.H., C.M., P.W.S.), Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, 75390; and Departments of Chemistry (S.H.K., J.A.K.), Food Science and Human Nutrition (Z.M.-E.), and Molecular and Integrative Physiology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801
| | - Jose Barrera
- Center for Pulmonary and Vascular Biology (K.L.C., J.B., M.U., J.U., L.H., C.M., P.W.S.), Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, 75390; and Departments of Chemistry (S.H.K., J.A.K.), Food Science and Human Nutrition (Z.M.-E.), and Molecular and Integrative Physiology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801
| | - Michihisa Umetani
- Center for Pulmonary and Vascular Biology (K.L.C., J.B., M.U., J.U., L.H., C.M., P.W.S.), Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, 75390; and Departments of Chemistry (S.H.K., J.A.K.), Food Science and Human Nutrition (Z.M.-E.), and Molecular and Integrative Physiology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801
| | - Junko Umetani
- Center for Pulmonary and Vascular Biology (K.L.C., J.B., M.U., J.U., L.H., C.M., P.W.S.), Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, 75390; and Departments of Chemistry (S.H.K., J.A.K.), Food Science and Human Nutrition (Z.M.-E.), and Molecular and Integrative Physiology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801
| | - Sung Hoon Kim
- Center for Pulmonary and Vascular Biology (K.L.C., J.B., M.U., J.U., L.H., C.M., P.W.S.), Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, 75390; and Departments of Chemistry (S.H.K., J.A.K.), Food Science and Human Nutrition (Z.M.-E.), and Molecular and Integrative Physiology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801
| | - Zeynep Madak-Erdogan
- Center for Pulmonary and Vascular Biology (K.L.C., J.B., M.U., J.U., L.H., C.M., P.W.S.), Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, 75390; and Departments of Chemistry (S.H.K., J.A.K.), Food Science and Human Nutrition (Z.M.-E.), and Molecular and Integrative Physiology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801
| | - Linzhang Huang
- Center for Pulmonary and Vascular Biology (K.L.C., J.B., M.U., J.U., L.H., C.M., P.W.S.), Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, 75390; and Departments of Chemistry (S.H.K., J.A.K.), Food Science and Human Nutrition (Z.M.-E.), and Molecular and Integrative Physiology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801
| | - Benita S Katzenellenbogen
- Center for Pulmonary and Vascular Biology (K.L.C., J.B., M.U., J.U., L.H., C.M., P.W.S.), Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, 75390; and Departments of Chemistry (S.H.K., J.A.K.), Food Science and Human Nutrition (Z.M.-E.), and Molecular and Integrative Physiology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801
| | - John A Katzenellenbogen
- Center for Pulmonary and Vascular Biology (K.L.C., J.B., M.U., J.U., L.H., C.M., P.W.S.), Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, 75390; and Departments of Chemistry (S.H.K., J.A.K.), Food Science and Human Nutrition (Z.M.-E.), and Molecular and Integrative Physiology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology (K.L.C., J.B., M.U., J.U., L.H., C.M., P.W.S.), Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, 75390; and Departments of Chemistry (S.H.K., J.A.K.), Food Science and Human Nutrition (Z.M.-E.), and Molecular and Integrative Physiology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology (K.L.C., J.B., M.U., J.U., L.H., C.M., P.W.S.), Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, 75390; and Departments of Chemistry (S.H.K., J.A.K.), Food Science and Human Nutrition (Z.M.-E.), and Molecular and Integrative Physiology (B.S.K.), University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801
| |
Collapse
|
72
|
Yokel RA. Physicochemical properties of engineered nanomaterials that influence their nervous system distribution and effects. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:2081-2093. [DOI: 10.1016/j.nano.2016.05.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 05/06/2016] [Accepted: 05/10/2016] [Indexed: 10/21/2022]
|
73
|
|
74
|
|
75
|
Nair M, Jayant RD, Kaushik A, Sagar V. Getting into the brain: Potential of nanotechnology in the management of NeuroAIDS. Adv Drug Deliv Rev 2016; 103:202-217. [PMID: 26944096 PMCID: PMC4935582 DOI: 10.1016/j.addr.2016.02.008] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 12/18/2022]
Abstract
In spite of significant advances in antiretroviral (ARV) therapy, the elimination of human immunodeficiency virus (HIV) reservoirs from the periphery and the central nervous system (CNS) remains a formidable task. The incapability of ARV to go across the blood-brain barrier (BBB) after systemic administration makes the brain one of the dominant HIV reservoirs. Thus, screening, monitoring, and elimination of HIV reservoirs from the brain remain a clinically daunting and key task. The practice and investigation of nanomedicine possesses potentials for therapeutics against neuroAIDS. This review highlights the advancements in nanoscience and nanotechnology to design and develop specific size therapeutic cargo for efficient navigation across BBB so as to recognize and eradicate HIV brain reservoirs. Different navigation and drug release strategies, their biocompatibility and efficacy with related challenges and future prospects are also discussed. This review would be an excellent platform to understand nano-enable multidisciplinary research to formulate efficient nanomedicine for the management of neuroAIDS.
Collapse
Key Words
- Anti-retroviral (ARV) therapy
- Blood–brain barrier (BBB)
- Bradykinin (PubChem CID: 439,201)
- CNS drug delivery
- Enfuvirtide (PubChem CID: 16,130,199), Lamivudine & Zidovudine (PubChem CID: 160,352)
- Ferrous oxide or iron (II) oxide (PubChem CID: 14,945)
- Foscarnet sodium (PubChem CID: 44,561)
- HIV monitoring
- HIV-1
- Magnetic nanoparticle
- Mannitol (PubChem CID: 6251)
- Nanotechnology
- Neopterin (PubChem CID: 4455)
- NeuroAIDS
- Pluronic-P85 (PubChem CID: 24,751)
- Saquinavir mesylate (PubChem CID: 60,934)
- Tenofovir disoproxil fumarate (PubChem CID: 6,398,764)
Collapse
Affiliation(s)
- Madhavan Nair
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA.
| | - Rahul Dev Jayant
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA.
| | - Ajeet Kaushik
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Vidya Sagar
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
76
|
Naoum GE, Tawadros F, Farooqi AA, Qureshi MZ, Tabassum S, Buchsbaum DJ, Arafat W. Role of nanotechnology and gene delivery systems in TRAIL-based therapies. Ecancermedicalscience 2016; 10:660. [PMID: 27594905 PMCID: PMC4990059 DOI: 10.3332/ecancer.2016.660] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Indexed: 12/11/2022] Open
Abstract
Since its identification as a member of the tumour necrosis factor (TNF) family, TRAIL (TNF-related apoptosis-inducing ligand) has emerged as a new avenue in apoptosis-inducing cancer therapies. Its ability to circumvent the chemoresistance of conventional therapeutics and to interact with cancer stem cells (CSCs) self-renewal pathways, amplified its potential as a cancer apoptotic agent. Many recombinant preparations of this death ligand and monoclonal antibodies targeting its death receptors have been tested in monotherapy and combinational clinical trials. Gene therapy is a new approach for cancer treatment which implies viral or non-viral functional transgene induction of apoptosis in cancer cells or repair of the underlying genetic abnormality on a molecular level. The role of this approach in overcoming the traditional barriers of radiation and chemotherapeutics systemic toxicity, risk of recurrence, and metastasis made it a promising platform for cancer treatment. The recent first Food Drug Administration (FDA) approved oncolytic herpes virus for melanoma treatment brings forth the potency of the cancer gene therapy approach in the future. Many gene delivery systems have been studied for intratumoural TRAIL gene delivery alone or in combination with chemotherapeutic agents to produce synergistic cancer cytotoxicity. However, there still remain many obstacles to be conquered for this different gene delivery systems. Nanomedicine on the other hand offers a new frontier for clinical trials and biomedical research. The FDA approved nanodrugs motivates horizon exploration for other nanoscale designed particles’ implications in gene delivery. In this review we aim to highlight the molecular role of TRAIL in apoptosis and interaction with cancer stem cells (CSCs) self-renewal pathways. Finally, we also aim to discuss the different roles of gene delivery systems, mesenchymal cells, and nanotechnology designs in TRAIL gene delivery.
Collapse
Affiliation(s)
| | - Fady Tawadros
- East Tennessee State University, 1276 Gilbreath Dr, Johnson City, TN 37604, USA
| | | | | | - Sobia Tabassum
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, Pakistan
| | - Donald J Buchsbaum
- University of Alabama at Birmingham, 1720 2nd Ave S, Birmingham, AL 35233, USA
| | - Waleed Arafat
- University of Alabama at Birmingham, 1720 2nd Ave S, Birmingham, AL 35233, USA; University of Alexandria, El-Gaish Rd, Egypt, Alexandria, Egypt
| |
Collapse
|
77
|
Self-assembled targeted nanoparticles based on transferrin-modified eight-arm-polyethylene glycol-dihydroartemisinin conjugate. Sci Rep 2016; 6:29461. [PMID: 27377918 PMCID: PMC4932499 DOI: 10.1038/srep29461] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/20/2016] [Indexed: 12/16/2022] Open
Abstract
Poor delivery of insoluble anticancer drugs has so far precluded their clinical application. In this study, an efficient tumor targeted-nanoparticle delivery system, transferrin-eight-arm-polyethylene glycol–dihydroartemisinin nanoparticles (TF-8arm-PEG-DHA NPs) for the vehiculation of dihydroartemisinin (DHA) was first prepared and evaluated for its targeting efficiency and cytotoxicity in vitro and in vivo to Lewis lung carcinoma (LLC) cells, which overexpress transferrin receptors (TFRs). The synthesized TF-8arm-PEG–DHA NPs had high solubility (~102 fold of free DHA), relatively high drug loading (~10 wt% DHA), long circulating half-life and moderate particle size (~147 nm). The in vitro cytotoxicity and in vivo tumor growth inhibition studies in LLC-tumor bearing mice confirmed the enhanced efficacy of TF-modified 8arm-PEG-DHA NPs compared to free DHA and non-modified 8arm-PEG-DHA NPs. All these results together supported that the formulation developed in this work exhibited great potential as an effective tumor targeting delivery system for insoluble anticancer drugs.
Collapse
|
78
|
Xu X, Li J, Han S, Tao C, Fang L, Sun Y, Zhu J, Liang Z, Li F. A novel doxorubicin loaded folic acid conjugated PAMAM modified with borneol, a nature dual-functional product of reducing PAMAM toxicity and boosting BBB penetration. Eur J Pharm Sci 2016; 88:178-90. [DOI: 10.1016/j.ejps.2016.02.015] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/24/2016] [Accepted: 02/26/2016] [Indexed: 12/20/2022]
|
79
|
Jayant RD, Sosa D, Kaushik A, Atluri V, Vashist A, Tomitaka A, Nair M. Current status of non-viral gene therapy for CNS disorders. Expert Opin Drug Deliv 2016; 13:1433-45. [PMID: 27249310 DOI: 10.1080/17425247.2016.1188802] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Viral and non-viral vectors have been used as methods of delivery in gene therapy for many CNS diseases. Currently, viral vectors such as adeno-associated viruses (AAV), retroviruses, lentiviruses, adenoviruses and herpes simplex viruses (HHV) are being used as successful vectors in gene therapy at clinical trial levels. However, many disadvantages have risen from their usage. Non-viral vectors like cationic polymers, cationic lipids, engineered polymers, nanoparticles, and naked DNA offer a much safer option and can therefore be explored for therapeutic purposes. AREAS COVERED This review discusses different types of viral and non-viral vectors for gene therapy and explores clinical trials for CNS diseases that have used these types of vectors for gene delivery. Highlights include non-viral gene delivery and its challenges, possible strategies to improve transfection, regulatory issues concerning vector usage, and future prospects for clinical applications. EXPERT OPINION Transfection efficiency of cationic lipids and polymers can be improved through manipulation of molecules used. Efficacy of cationic lipids is dependent on cationic charge, saturation levels, and stability of linkers. Factors determining efficacy of cationic polymers are total charge density, molecular weights, and complexity of molecule. All of the above mentioned parameters must be taken care for efficient gene delivery.
Collapse
Affiliation(s)
- Rahul Dev Jayant
- a Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine , Florida International University , Miami , FL , USA
| | - Daniela Sosa
- a Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine , Florida International University , Miami , FL , USA
| | - Ajeet Kaushik
- a Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine , Florida International University , Miami , FL , USA
| | - Venkata Atluri
- a Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine , Florida International University , Miami , FL , USA
| | - Arti Vashist
- a Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine , Florida International University , Miami , FL , USA
| | - Asahi Tomitaka
- a Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine , Florida International University , Miami , FL , USA
| | - Madhavan Nair
- a Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine , Florida International University , Miami , FL , USA
| |
Collapse
|
80
|
Gomes MJ, Fernandes C, Martins S, Borges F, Sarmento B. Tailoring Lipid and Polymeric Nanoparticles as siRNA Carriers towards the Blood-Brain Barrier - from Targeting to Safe Administration. J Neuroimmune Pharmacol 2016; 12:107-119. [PMID: 27209050 DOI: 10.1007/s11481-016-9685-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/09/2016] [Indexed: 01/25/2023]
Abstract
Blood-brain barrier is a tightly packed layer of endothelial cells surrounding the brain that acts as the main obstacle for drugs enter the central nervous system (CNS), due to its unique features, as tight junctions and drug efflux systems. Therefore, since the incidence of CNS disorders is increasing worldwide, medical therapeutics need to be improved. Consequently, aiming to surpass blood-brain barrier and overcome CNS disabilities, silencing P-glycoprotein as a drug efflux transporter at brain endothelial cells through siRNA is considered a promising approach. For siRNA enzymatic protection and efficient delivery to its target, two different nanoparticles platforms, solid lipid (SLN) and poly-lactic-co-glycolic (PLGA) nanoparticles were used in this study. Polymeric PLGA nanoparticles were around 115 nm in size and had 50 % of siRNA association efficiency, while SLN presented 150 nm and association efficiency close to 52 %. Their surface was functionalized with a peptide-binding transferrin receptor, in a site-oriented manner confirmed by NMR, and their targeting ability against human brain endothelial cells was successfully demonstrated by fluorescence microscopy and flow cytometry. The interaction of modified nanoparticles with brain endothelial cells increased 3-fold compared to non-modified lipid nanoparticles, and 4-fold compared to non-modified PLGA nanoparticles, respectively. These nanosystems, which were also demonstrated to be safe for human brain endothelial cells, without significant cytotoxicity, bring a new hopeful breath to the future of brain diseases therapies.
Collapse
Affiliation(s)
- Maria João Gomes
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- INEB, Instituto de Engenharia Biomédica, Biocarrier Group, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- ICBAS, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Carlos Fernandes
- CIQUP/Departamento de Química, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, 687, 4169-007, Porto, Portugal
| | - Susana Martins
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, -5230, Odense, DK, Denmark
| | - Fernanda Borges
- CIQUP/Departamento de Química, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, 687, 4169-007, Porto, Portugal
| | - Bruno Sarmento
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.
- INEB, Instituto de Engenharia Biomédica, Biocarrier Group, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.
- Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, CESPU, Rua Central de Gandra, 1317, 4585-116, Gandra, Portugal.
| |
Collapse
|
81
|
Zeng Y, Kurokawa Y, Zeng Q, Win-Shwe TT, Nansai H, Zhang Z, Sone H. Effects of Polyamidoamine Dendrimers on a 3-D Neurosphere System Using Human Neural Progenitor Cells. Toxicol Sci 2016; 152:128-44. [DOI: 10.1093/toxsci/kfw068] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
82
|
Kafa H, Wang JTW, Rubio N, Klippstein R, Costa PM, Hassan HAFM, Sosabowski JK, Bansal SS, Preston JE, Abbott NJ, Al-Jamal KT. Translocation of LRP1 targeted carbon nanotubes of different diameters across the blood-brain barrier in vitro and in vivo. J Control Release 2016; 225:217-29. [PMID: 26809004 PMCID: PMC4778610 DOI: 10.1016/j.jconrel.2016.01.031] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 01/15/2016] [Accepted: 01/18/2016] [Indexed: 01/18/2023]
Abstract
Brain glioblastoma and neurodegenerative diseases are still largely untreated due to the inability of most drugs to cross the blood-brain barrier (BBB). Nanoparticles have emerged as promising tools for drug delivery applications to the brain; in particular carbon nanotubes (CNTs) that have shown an intrinsic ability to cross the BBB in vitro and in vivo. Angiopep-2 (ANG), a ligand for the low-density lipoprotein receptor-related protein-1 (LRP1), has also shown promising results as a targeting ligand for brain delivery using nanoparticles (NPs). Here, we investigate the ability of ANG-targeted chemically-functionalised multi-walled carbon nanotubes (f-MWNTs) to cross the BBB in vitro and in vivo. ANG was conjugated to wide and thin f-MWNTs creating w-MWNT-ANG and t-MWNT-ANG, respectively. All f-MWNTs were radiolabelled to facilitate quantitative analyses by γ-scintigraphy. ANG conjugation to f-MWNTs enhanced BBB transport of w- and t-MWNTs-ANG compared to their non-targeted equivalents using an in vitro co-cultured BBB model consisting of primary porcine brain endothelial cells (PBEC) and primary rat astrocytes. Additionally, following intravenous administration w-MWNTs-ANG showed significantly higher whole brain uptake than the non-targeted w-MWNT in vivo reaching ~2% injected dose per g of brain (%ID/g) within the first hour post-injection. Furthermore, using a syngeneic glioma model, w-MWNT-ANG showed enhanced uptake in glioma brain compared to normal brain at 24h post-injection. t-MWNTs-ANG, on the other hand, showed higher brain accumulation than w-MWNTs. However, no significant differences were observed between t-MWNT and t-MWNT-ANG indicating the importance of f-MWNTs diameter towards their brain accumulation. The inherent brain accumulation ability of f-MWNTs coupled with improved brain-targeting by ANG favours the future clinical applications of f-MWNT-ANG to deliver active therapeutics for brain glioma therapy.
Collapse
Affiliation(s)
- Houmam Kafa
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Julie Tzu-Wen Wang
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Noelia Rubio
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Rebecca Klippstein
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Pedro M Costa
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Hatem A F M Hassan
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Jane K Sosabowski
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Sukhvinder S Bansal
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Jane E Preston
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - N Joan Abbott
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Khuloud T Al-Jamal
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK.
| |
Collapse
|
83
|
Zhao C, Shao L, Lu J, Deng X, Wu Y. Tumor Acidity-Induced Sheddable Polyethylenimine-Poly(trimethylene carbonate)/DNA/Polyethylene Glycol-2,3-Dimethylmaleicanhydride Ternary Complex for Efficient and Safe Gene Delivery. ACS APPLIED MATERIALS & INTERFACES 2016; 8:6400-6410. [PMID: 26904916 DOI: 10.1021/acsami.6b00825] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Amphiphilic PEI derivatives/DNA complexes are widely used for DNA delivery, but they are unstable in vivo and have cytotoxicity due to the excess cationic charge. PEGylation of cationic complexes can improve sterical stability and biocompatibility. However, PEGylation significantly inhibits cellular uptake and endosomal escape. In this work, sheddable ternary complexes were developed by coating a tumor acidity-sensitive β-carboxylic amide functionalized PEG layer on the binary complexes of amphiphilic cationic polyethylenimine-poly(trimethylene carbonate) nanoparticles/DNA (PEI-PTMC/DNA). Such sheddable ternary complexes markedly reduced their nonspecific interactions with serum protein in the bloodstream and obtained minimal cytotoxicity due to the protection of the PEG shell. At the tumor site, the PEG layer was deshielded by responding to the tumor acidic microenvironment and the positively charged complexes re-exposed that had higher affinity with negatively charged cell membranes. Meanwhile the positively charged complexes facilitated endosomal escape. Accordingly, this delivery system improved the biocompatibility of gene-loaded complexes and enhanced the gene transfection efficiency. Such PEGylated complexes with the ability to deshield the PEG layer at the target tissues hold great promise for efficient and safe gene delivery in vivo.
Collapse
Affiliation(s)
- Caiyan Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Leihou Shao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Jianqing Lu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Xiongwei Deng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| | - Yan Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China
| |
Collapse
|
84
|
Zoetebier B, Sohrabi A, Lou B, Hempenius MA, Hennink WE, Vancso GJ. PEG stabilized DNA – poly(ferrocenylsilane) polyplexes for gene delivery. Chem Commun (Camb) 2016; 52:7707-10. [DOI: 10.1039/c6cc02733d] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Polycationic poly(ferrocenylsilane)s (PFS) with tunable amounts of PEG side chains were used for the condensation of DNA into polyplexes of 110 nm in 5.0 mM HEPES.
Collapse
Affiliation(s)
- B. Zoetebier
- Department of Materials Science and Technology of Polymers
- MESA+ Institute for Nanotechnology
- University of Twente
- 7500 AE Enschede
- The Netherlands
| | - A. Sohrabi
- Department of Materials Science and Technology of Polymers
- MESA+ Institute for Nanotechnology
- University of Twente
- 7500 AE Enschede
- The Netherlands
| | - B. Lou
- Department of Pharmaceutics
- Utrecht Institute for Pharmaceutical Sciences
- Utrecht University
- 3584 CG Utrecht
- The Netherlands
| | - M. A. Hempenius
- Department of Materials Science and Technology of Polymers
- MESA+ Institute for Nanotechnology
- University of Twente
- 7500 AE Enschede
- The Netherlands
| | - W. E. Hennink
- Department of Pharmaceutics
- Utrecht Institute for Pharmaceutical Sciences
- Utrecht University
- 3584 CG Utrecht
- The Netherlands
| | - G. J. Vancso
- Department of Materials Science and Technology of Polymers
- MESA+ Institute for Nanotechnology
- University of Twente
- 7500 AE Enschede
- The Netherlands
| |
Collapse
|
85
|
Hou W, Wei P, Kong L, Guo R, Wang S, Shi X. Partially PEGylated dendrimer-entrapped gold nanoparticles: a promising nanoplatform for highly efficient DNA and siRNA delivery. J Mater Chem B 2016; 4:2933-2943. [DOI: 10.1039/c6tb00710d] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Partially PEGylated dendrimer-entrapped gold nanoparticles can be used as a promising nanoplatform for highly efficient pDNA and siRNA delivery.
Collapse
Affiliation(s)
- Wenxiu Hou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Chemistry
- Chemical Engineering and Biotechnology
- Donghua University
- Shanghai 201620
| | - Ping Wei
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Chemistry
- Chemical Engineering and Biotechnology
- Donghua University
- Shanghai 201620
| | - Lingdan Kong
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Chemistry
- Chemical Engineering and Biotechnology
- Donghua University
- Shanghai 201620
| | - Rui Guo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Chemistry
- Chemical Engineering and Biotechnology
- Donghua University
- Shanghai 201620
| | - Shige Wang
- College of Science
- University of Shanghai for Science and Technology
- Shanghai 200093
- People's Republic of China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Chemistry
- Chemical Engineering and Biotechnology
- Donghua University
- Shanghai 201620
| |
Collapse
|
86
|
Gao S, Li J, Jiang C, Hong B, Hao B. Plasmid pORF-hTRAIL targeting to glioma using transferrin-modified polyamidoamine dendrimer. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 10:1-11. [PMID: 26719669 PMCID: PMC4690643 DOI: 10.2147/dddt.s95843] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A gene drug delivery system for glioma therapy based on transferrin (Tf)-modified polyamidoamine dendrimer (PAMAM) was prepared. Gene drug, tumor necrosis factor-related apoptosis-inducing ligand (hTRAIL)-encoding plasmid open reading frame (pORF-hTRAIL, Trail), was condensed by Tf-modified PAMAM to form nanoparticles (NPs). PAMAM-PEG-Tf/DNA NPs showed higher cellular uptake, in vitro gene expression, and cytotoxicity than PAMAM-PEG/DNA NPs in C6 cells. The in vivo targeting efficacy of NPs was visualized by ex vivo fluorescence imaging. Tf-modified NPs showed obvious glioma-targeting trend. Plasmid encoding green fluorescence protein (GFP) was also condensed by modified or unmodified PAMAM to evaluate the in vivo gene expression level. The PAMAM-PEG-Tf/plasmid encoding enhanced green fluorescence protein (pEGFP) NPs exhibited higher GFP expression level than PAMAM-PEG/pEGFP NPs. TUNEL assay revealed that Tf-modified NPs could induce much more tumor apoptosis. The median survival time of PAMAM-PEG-Tf/Trail-treated rats (28.5 days) was longer than that of rats treated with PAMAM-PEG/Trail (25.5 days), temozolomide (24.5 days), PAMAM-PEG-Tf/pEGFP (19 days), or saline (17 days). The therapeutic effect was further confirmed by magnetic resonance imaging. This study demonstrated that targeting gene delivery system had potential application for the treatment of glioma.
Collapse
Affiliation(s)
- Song Gao
- Department of Clinical Laboratory, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jianfeng Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Bo Hong
- Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Bing Hao
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| |
Collapse
|
87
|
Lukowiak MC, Thota BN, Haag R. Dendritic core–shell systems as soft drug delivery nanocarriers. Biotechnol Adv 2015; 33:1327-41. [DOI: 10.1016/j.biotechadv.2015.03.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 03/19/2015] [Accepted: 03/22/2015] [Indexed: 12/29/2022]
|
88
|
Evaluation of improved PAMAM-G5 conjugates for gene delivery targeted to the transferrin receptor. Eur J Pharm Biopharm 2015; 94:116-22. [DOI: 10.1016/j.ejpb.2015.05.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 03/06/2015] [Accepted: 05/07/2015] [Indexed: 11/18/2022]
|
89
|
Dehshahri A, Sadeghpour H. Surface decorations of poly(amidoamine) dendrimer by various pendant moieties for improved delivery of nucleic acid materials. Colloids Surf B Biointerfaces 2015; 132:85-102. [PMID: 26022400 DOI: 10.1016/j.colsurfb.2015.05.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 05/05/2015] [Accepted: 05/07/2015] [Indexed: 12/22/2022]
|
90
|
Somani S, Dufès C. Applications of dendrimers for brain delivery and cancer therapy. Nanomedicine (Lond) 2015; 9:2403-14. [PMID: 25413857 DOI: 10.2217/nnm.14.130] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Dendrimers are emerging as potential nonviral vectors for the efficient delivery of drugs and nucleic acids to the brain and cancer cells. These polymers are highly branched, 3D macromolecules with modifiable surface functionalities and available internal cavities that make them attractive as delivery systems for drug and gene delivery applications. This article highlights the recent therapeutic advances resulting from the use of dendrimers for brain targeting and cancer treatment.
Collapse
Affiliation(s)
- Sukrut Somani
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | | |
Collapse
|
91
|
Zarebkohan A, Najafi F, Moghimi HR, Hemmati M, Deevband MR, Kazemi B. Synthesis and characterization of a PAMAM dendrimer nanocarrier functionalized by SRL peptide for targeted gene delivery to the brain. Eur J Pharm Sci 2015; 78:19-30. [PMID: 26118442 DOI: 10.1016/j.ejps.2015.06.024] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/17/2015] [Accepted: 06/25/2015] [Indexed: 12/22/2022]
Abstract
Blood-brain barrier inhibits most of drugs and genetic materials from reaching the brain. So, developing high efficiency carriers for gene and drug delivery to the brain, is the challenging area in pharmaceutical sciences. This investigation aimed to target DNA to brain using Serine-Arginine-Leucine (SRL) functionalized PAMAM dendrimers as a novel gene delivery system. The SRL peptide was linked on G4 PAMAM dendrimers using bifunctional PEG. DNA was then loaded in these functionalized nanoparticles and their physicochemical properties and cellular uptake/distribution evaluated by AFM, NMR, FTIR and fluorescence and confocal microscopy. Also, biodistribution and brain localization of nanoparticles were studied after IV injection of nanoparticles into rat tail. Unmodified nanoparticles were used as control in all evaluations. In vitro studies showed that SRL-modified nanoparticles have good transfection efficacy and low toxicity. Results also showed that SRL is a LRP ligand and SRL-modified nanoparticles internalized by clathrin/caveolin energy-dependent endocytosis to brain capillary endothelial cells. After intravenous administration, the SRL-modified nanoparticles were able to cross the blood-brain barrier and enter the brain parenchyma. Our result showed that, SRL-modified nanoparticles provide a safe and effective nanocarrier for brain gene delivery.
Collapse
Affiliation(s)
- Amir Zarebkohan
- Biomedical Engineering and Medical Physics Department, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farhood Najafi
- Department of Resin and Additives, Institute for Color Science and Technology, Tehran, Iran
| | - Hamid Reza Moghimi
- School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hemmati
- Biomedical Engineering and Medical Physics Department, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Deevband
- Biomedical Engineering and Medical Physics Department, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahram Kazemi
- Department of Biotechnology, Faculty of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
92
|
Edagwa BJ, Zhou T, McMillan JM, Liu XM, Gendelman HE. Development of HIV reservoir targeted long acting nanoformulated antiretroviral therapies. Curr Med Chem 2015; 21:4186-98. [PMID: 25174930 DOI: 10.2174/0929867321666140826114135] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 08/19/2014] [Accepted: 08/22/2014] [Indexed: 11/22/2022]
Abstract
Human immunodeficiency virus (HIV) infection commonly results in a myriad of comorbid conditions secondary to immune deficiency. Infection also affects broad organ system function. Although current antiretroviral therapy (ART) reduces disease morbidity and mortality through effective control of peripheral viral load, restricted infection in HIV reservoirs including gut, lymphoid and central nervous system tissues, is not eliminated. What underlies these events is, in part, poor ART penetrance into each organ across tissue barriers, viral mutation and the longevity of infected cells. We posit that one means to improve these disease outcomes is through nanotechnology. To this end, this review discusses a broad range of cutting-edge nanomedicines and nanomedicine platforms that are or can be used to improve ART delivery. Discussion points include how polymer-drug conjugates, dendrimers, micelles, liposomes, solid lipid nanoparticles and polymeric nanoparticles can be harnessed to best yield cell-based delivery systems. When completely developed, such nanomedicine platforms have the potential to clear reservoirs of viral infection.
Collapse
Affiliation(s)
| | | | | | | | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
93
|
Abstract
In this article, advances in designing polymeric nanoparticles for targeted cancer gene therapy are reviewed. Characterization and evaluation of biomaterials, targeting ligands, and transcriptional elements are each discussed. Advances in biomaterials have driven improvements to nanoparticle stability and tissue targeting, conjugation of ligands to the surface of polymeric nanoparticles enable binding to specific cancer cells, and the design of transcriptional elements has enabled selective DNA expression specific to the cancer cells. Together, these features have improved the performance of polymeric nanoparticles as targeted non-viral gene delivery vectors to treat cancer. As polymeric nanoparticles can be designed to be biodegradable, non-toxic, and to have reduced immunogenicity and tumorigenicity compared to viral platforms, they have significant potential for clinical use. Results of polymeric gene therapy in clinical trials and future directions for the engineering of nanoparticle systems for targeted cancer gene therapy are also presented.
Collapse
Affiliation(s)
- Jayoung Kim
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David R. Wilson
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Camila G. Zamboni
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Jordan J. Green
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
94
|
Affiliation(s)
- Preeti Kumari
- Department of Pharmacy, Birla Institute of Technology and Science – Pilani, Hyderabad Campus, Hyderabad, Andhra Pradesh, India
| | - Balaram Ghosh
- Department of Pharmacy, Birla Institute of Technology and Science – Pilani, Hyderabad Campus, Hyderabad, Andhra Pradesh, India
| | - Swati Biswas
- Department of Pharmacy, Birla Institute of Technology and Science – Pilani, Hyderabad Campus, Hyderabad, Andhra Pradesh, India
| |
Collapse
|
95
|
Yang J, Zhang Q, Chang H, Cheng Y. Surface-Engineered Dendrimers in Gene Delivery. Chem Rev 2015; 115:5274-300. [PMID: 25944558 DOI: 10.1021/cr500542t] [Citation(s) in RCA: 325] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Jiepin Yang
- Shanghai
Key Laboratory of
Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, P. R. China
| | - Qiang Zhang
- Shanghai
Key Laboratory of
Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, P. R. China
| | - Hong Chang
- Shanghai
Key Laboratory of
Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, P. R. China
| | - Yiyun Cheng
- Shanghai
Key Laboratory of
Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, P. R. China
| |
Collapse
|
96
|
Gomes MJ, Martins S, Sarmento B. siRNA as a tool to improve the treatment of brain diseases: Mechanism, targets and delivery. Ageing Res Rev 2015; 21:43-54. [PMID: 25796492 DOI: 10.1016/j.arr.2015.03.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 03/10/2015] [Accepted: 03/16/2015] [Indexed: 10/23/2022]
Abstract
As the population ages, brain pathologies such as neurodegenerative diseases and brain cancer increase their incidence, being the need to find successful treatments of upmost importance. Drug delivery to the central nervous system (CNS) is required in order to reach diseases causes and treat them. However, biological barriers, mainly blood-brain barrier (BBB), are the key obstacles that prevent the effectiveness of possible treatments due to their ability to strongly limit the perfusion of compounds into the brain. Over the past decades, new approaches towards overcoming BBB and its efflux transporters had been proposed. One of these approaches here reviewed is through small interfering RNA (siRNA), which is capable to specifically target one gene and silence it in a post-transcriptional way. There are different possible functional proteins at the BBB, as the ones responsible for transport or just for its tightness, which could be a siRNA target. As important as the effective silence is the way to delivery siRNA to its anatomical site of action. This is where nanotechnology-based systems may help, by protecting siRNA circulation and providing cell/tissue-targeting and intracellular siRNA delivery. After an initial overview on incidence of brain diseases and basic features of the CNS, BBB and its efflux pumps, this review focuses on recent strategies to reach brain based on siRNA, and how to specifically target these approaches in order to treat brain diseases.
Collapse
|
97
|
Lächelt U, Wagner E. Nucleic Acid Therapeutics Using Polyplexes: A Journey of 50 Years (and Beyond). Chem Rev 2015; 115:11043-78. [DOI: 10.1021/cr5006793] [Citation(s) in RCA: 469] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Ulrich Lächelt
- Pharmaceutical
Biotechnology, Department of Pharmacy, Ludwig Maximilians Universität, 81377 Munich, Germany
- Nanosystems
Initiative
Munich (NIM), 80799 Munich, Germany
| | - Ernst Wagner
- Pharmaceutical
Biotechnology, Department of Pharmacy, Ludwig Maximilians Universität, 81377 Munich, Germany
- Nanosystems
Initiative
Munich (NIM), 80799 Munich, Germany
| |
Collapse
|
98
|
Kong L, Alves CS, Hou W, Qiu J, Möhwald H, Tomás H, Shi X. RGD peptide-modified dendrimer-entrapped gold nanoparticles enable highly efficient and specific gene delivery to stem cells. ACS APPLIED MATERIALS & INTERFACES 2015; 7:4833-4843. [PMID: 25658033 DOI: 10.1021/am508760w] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
We report the use of arginine-glycine-aspartic (Arg-Gly-Asp, RGD) peptide-modified dendrimer-entrapped gold nanoparticles (Au DENPs) for highly efficient and specific gene delivery to stem cells. In this study, generation 5 poly(amidoamine) dendrimers modified with RGD via a poly(ethylene glycol) (PEG) spacer and with PEG monomethyl ether were used as templates to entrap gold nanoparticles (AuNPs). The native and the RGD-modified PEGylated dendrimers and the respective well characterized Au DENPs were used as vectors to transfect human mesenchymal stem cells (hMSCs) with plasmid DNA (pDNA) carrying both the enhanced green fluorescent protein and the luciferase (pEGFPLuc) reporter genes, as well as pDNA encoding the human bone morphogenetic protein-2 (hBMP-2) gene. We show that all vectors are capable of transfecting the hMSCs with both pDNAs. Gene transfection using pEGFPLuc was demonstrated by quantitative Luc activity assay and qualitative evaluation by fluorescence microscopy. For the transfection with hBMP-2, the gene delivery efficiency was evaluated by monitoring the hBMP-2 concentration and the level of osteogenic differentiation of the hMSCs via alkaline phosphatase activity, osteocalcin secretion, calcium deposition, and von Kossa staining assays. Our results reveal that the stem cell gene delivery efficiency is largely dependent on the composition and the surface functionality of the dendrimer-based vectors. The coexistence of RGD and AuNPs rendered the designed dendrimeric vector with specific stem cell binding ability likely via binding of integrin receptor on the cell surface and improved three-dimensional conformation of dendrimers, which is beneficial for highly efficient and specific stem cell gene delivery applications.
Collapse
Affiliation(s)
- Lingdan Kong
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University , Shanghai 201620, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
99
|
Alajangi HK, Santhiya D. Fluorescence and Förster resonance energy transfer investigations on DNA oligonucleotide and PAMAM dendrimer packing interactions in dendriplexes. Phys Chem Chem Phys 2015; 17:8680-91. [PMID: 25738189 DOI: 10.1039/c4cp05295a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Considering the importance of short oligonucleotide packing in dendriplex-mediated gene delivery, a direct insight into the 14-mer oligonucleotide and dendrimer interactions using fluorescence and FRET techniques is the focus of this study. Fluorometric titrations of various fluorophore-tagged oligonucleotides with the first three PAMAM dendrimer generations showed a decrease in the fluorescence intensity with two break points, namely Z and Z, for each titration. The first break point for each dendrimer was identical to the neutralization point observed by basic biophysical studies for the corresponding dendrimer generations. Additionally, FRET studies on dual tagged oligonucleotide (DFT) molecules revealed a third break point at the charge ratio (Z) where there was the highest fluorescence energy transfer from the donor to the acceptor fluorophores. Altogether, dendriplex formation was considered to take place via three steps with an increase in the dendrimer concentration, where initially there was monomeric complexation at the neutralization point (Z) followed by loosely held molecular aggregation of the dendrimer (Z). In the final step, dendrimer molecular aggregates were held tightly together for the closest possible packing of the oligonucleotide molecules onto their surface. The effective molecular packing is identified by the highest FRET intensity for the dendrimer of generation 2 at a charge ratio of 0.34 (Z±).
Collapse
Affiliation(s)
- Hema Kumari Alajangi
- Department of Applied Chemistry and Polymer Technology, Delhi Technological University, Bawana Road, Delhi-110 042, India.
| | | |
Collapse
|
100
|
Yang Z, Gao D, Cao Z, Zhang C, Cheng D, Liu J, Shuai X. Drug and gene co-delivery systems for cancer treatment. Biomater Sci 2015. [PMID: 26221938 DOI: 10.1039/c4bm00369a] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cancer remains a major killer and a leading cause of death in the world; thus, a growing number of new treatments have been focused on cancer therapy over the past few decades. Chemotherapy, which is thought to be a powerful strategy for cancer treatment, has been widely used in clinical therapy in recent years. However, due to the complexity of cancer, a single therapeutic approach is insufficient for the suppression of cancer growth and migration. Therefore, increasing attention has been paid to the use of smart multifunctional carriers and combinatorially delivers chemotherapeutic drugs and functional genes in order to maximize therapeutic efficiency. Combination therapy using selected drugs and genes can not only overcome multidrug resistance and inhibit the cellular anti-apoptotic process but also achieve a synergistic therapeutic effect. Because multifunctional nanocarriers are important for achieving these goals, this review will illustrate and discuss some advanced biomaterial nanocarriers for co-delivering therapeutic genes and drugs, including multifunctional micelles, liposomes, polymeric conjugates and inorganic nanoparticles. In addition, the challenges and future perspectives for co-delivery systems, containing therapeutic drugs and genes to achieve better therapeutic effects for cancer treatment will be discussed.
Collapse
Affiliation(s)
- Zhe Yang
- Department of Biomedical Engineering, School of Engineering, Sun Yat-sen University, Guangzhou, Guangdong 510006, China.
| | | | | | | | | | | | | |
Collapse
|