1
|
Viltres H, López YC, Leyva C, Gupta NK, Naranjo AG, Acevedo–Peña P, Sanchez-Diaz A, Bae J, Kim KS. Polyamidoamine dendrimer-based materials for environmental applications: A review. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.116017] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
2
|
Maurya A, Singh AK, Mishra G, Kumari K, Rai A, Sharma B, Kulkarni GT, Awasthi R. Strategic use of nanotechnology in drug targeting and its consequences on human health: A focused review. Interv Med Appl Sci 2019; 11:38-54. [PMID: 32148902 PMCID: PMC7044564 DOI: 10.1556/1646.11.2019.04] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 01/03/2019] [Accepted: 01/28/2019] [Indexed: 02/06/2023] Open
Abstract
Since the development of first lipid-based nanocarrier system, about 15% of the present pharmaceutical market uses nanomedicines to achieve medical benefits. Nanotechnology is an advanced area to meliorate the delivery of compounds for improved medical diagnosis and curing disease. Nanomedicines are gaining significant interest due to the ultra small size and large surface area to mass ratio. In this review, we discuss the potential of nanotechnology in delivering of active moieties for the disease therapy including their toxicity evidences. This communication will help the formulation scientists in understanding and exploring the new aspects of nanotechnology in the field of nanomedicine.
Collapse
Affiliation(s)
- Anand Maurya
- Faculty of Ayurveda, Department of Medicinal Chemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Anurag Kumar Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Gaurav Mishra
- Faculty of Ayurveda, Department of Medicinal Chemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Komal Kumari
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Ajmer, India
| | - Arati Rai
- Department of Pharmacy, Hygia Institute of Pharmaceutical Education and Research, Lucknow, India
| | - Bhupesh Sharma
- Amity Institute of Pharmacy, Amity University, Noida, India
| | | | | |
Collapse
|
3
|
Parboosing R, Chonco L, de la Mata FJ, Govender T, Maguire GE, Kruger HG. Potential inhibition of HIV-1 encapsidation by oligoribonucleotide-dendrimer nanoparticle complexes. Int J Nanomedicine 2017; 12:317-325. [PMID: 28115849 PMCID: PMC5221794 DOI: 10.2147/ijn.s114446] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background Encapsidation, the process during which the genomic RNA of HIV is packaged into viral particles, is an attractive target for antiviral therapy. This study explores a novel nanotechnology-based strategy to inhibit HIV encapsidation by an RNA decoy mechanism. The design of the 16-mer oligoribonucleotide (RNA) decoy is based on the sequence of stem loop 3 (SL3) of the HIV packaging signal (Ψ). Recognition of the packaging signal is essential to the encapsidation process. It is theorized that the decoy RNA, by mimicking the packaging signal, will disrupt HIV packaging if efficiently delivered into lymphocytes by complexation with a carbosilane dendrimer. The aim of the study is to measure the uptake, toxicity, and antiviral activity of the dendrimer–RNA nanocomplex. Materials and methods A dendriplex was formed between cationic carbosilane dendrimers and the RNA decoy. Uptake of the fluorescein-labeled RNA into MT4 lymphocytes was determined by flow cytometry and confocal microscopy. The cytoprotective effect (50% effective concentration [EC50]) and the effect on HIV replication were determined in vitro by the methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay and viral load measurements, respectively. Results Flow cytometry and confocal imaging demonstrated efficient transfection of lymphocytes. The dendriplex containing the Ψ decoy showed some activity (EC50 =3.20 µM, selectivity index =8.4). However, there was no significant suppression of HIV viral load. Conclusion Oligoribonucleotide decoys containing SL3 of the packaging sequence are efficiently delivered into lymphocytes by carbosilane dendrimers where they exhibit a modest cytoprotective effect against HIV infection.
Collapse
Affiliation(s)
- Raveen Parboosing
- Department of Virology, University of KwaZulu-Natal; National Health Laboratory Service, Durban, South Africa
| | - Louis Chonco
- Department of Virology, University of KwaZulu-Natal; National Health Laboratory Service, Durban, South Africa
| | - Francisco Javier de la Mata
- Organic and Inorganic Chemistry Department, University of Alcalá, Alcalá de Henares; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Thavendran Govender
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Durban, South Africa
| | - Glenn Em Maguire
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Durban, South Africa
| | - Hendrik G Kruger
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
4
|
Márquez-Miranda V, Peñaloza JP, Araya-Durán I, Reyes R, Vidaurre S, Romero V, Fuentes J, Céric F, Velásquez L, González-Nilo FD, Otero C. Effect of Terminal Groups of Dendrimers in the Complexation with Antisense Oligonucleotides and Cell Uptake. NANOSCALE RESEARCH LETTERS 2016; 11:66. [PMID: 26847692 PMCID: PMC4742457 DOI: 10.1186/s11671-016-1260-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 01/09/2016] [Indexed: 06/05/2023]
Abstract
Poly(amidoamine) dendrimers are the most recognized class of dendrimer. Amino-terminated (PAMAM-NH2) and hydroxyl-terminated (PAMAM-OH) dendrimers of generation 4 are widely used, since they are commercially available. Both have different properties, mainly based on their different overall charges at physiological pH. Currently, an important function of dendrimers as carriers of short single-stranded DNA has been applied. These molecules, known as antisense oligonucleotides (asODNs), are able to inhibit the expression of a target mRNA. Whereas PAMAM-NH2 dendrimers have shown to be able to transfect plasmid DNA, PAMAM-OH dendrimers have not shown the same successful results. However, little is known about their interaction with shorter and more flexible molecules such as asODNs. Due to several initiatives, the use of these neutral dendrimers as a scaffold to introduce other functional groups has been proposed. Because of its low cytotoxicity, it is relevant to understand the molecular phenomena involving these types of dendrimers. In this work, we studied the behavior of an antisense oligonucleotide in presence of both types of dendrimers using molecular dynamics simulations, in order to elucidate if they are able to form stable complexes. In this manner, we demonstrated at atomic level that PAMAM-NH2, unlike PAMAM-OH, could form a well-compacted complex with asODN, albeit PAMAM-OH can also establish stable interactions with the oligonucleotide. The biological activity of asODN in complex with PAMAM-NH2 dendrimer was also shown. Finally, we revealed that in contact with PAMAM-OH, asODN remains outside the cells as TIRF microscopy results showed, due to its poor interaction with this dendrimer and cell membranes.
Collapse
Affiliation(s)
- Valeria Márquez-Miranda
- Facultad de Biología, Center for Bioinformatics and Integrative Biology (CBIB), Universidad Andres Bello, Republica 239, Santiago, Chile
- Fundación Fraunhofer Chile Research, M. Sánchez Fontecilla 310 piso 14, Las Condes, Chile
| | - Juan Pablo Peñaloza
- Facultad de Medicina, Center for Integrative Medicine and Innovative Science, Universidad Andres Bello, Santiago, Chile
| | - Ingrid Araya-Durán
- Facultad de Biología, Center for Bioinformatics and Integrative Biology (CBIB), Universidad Andres Bello, Republica 239, Santiago, Chile
- Fundación Fraunhofer Chile Research, M. Sánchez Fontecilla 310 piso 14, Las Condes, Chile
| | - Rodrigo Reyes
- Facultad de Medicina, Center for Integrative Medicine and Innovative Science, Universidad Andres Bello, Santiago, Chile
| | - Soledad Vidaurre
- Departamento Ciencias Químicas y Biológicas, Laboratorio de Bionanotecnología, Universidad Bernardo O'Higgins, Santiago, Chile
| | - Valentina Romero
- Departamento Ciencias Químicas y Biológicas, Laboratorio de Bionanotecnología, Universidad Bernardo O'Higgins, Santiago, Chile
| | - Juan Fuentes
- Facultad de Biología, Laboratorio de Microbiología, Universidad Andres Bello, Republica 217, Santiago, Chile
| | - Francisco Céric
- Laboratorio de Neurociencias Cognitivas, Facultad de Psicología, Universidad del Desarrollo, Santiago, Chile
| | - Luis Velásquez
- Fundación Fraunhofer Chile Research, M. Sánchez Fontecilla 310 piso 14, Las Condes, Chile
- Facultad de Medicina, Center for Integrative Medicine and Innovative Science, Universidad Andres Bello, Santiago, Chile
| | - Fernando D González-Nilo
- Facultad de Biología, Center for Bioinformatics and Integrative Biology (CBIB), Universidad Andres Bello, Republica 239, Santiago, Chile.
- Fundación Fraunhofer Chile Research, M. Sánchez Fontecilla 310 piso 14, Las Condes, Chile.
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.
| | - Carolina Otero
- Fundación Fraunhofer Chile Research, M. Sánchez Fontecilla 310 piso 14, Las Condes, Chile.
- Facultad de Medicina, Center for Integrative Medicine and Innovative Science, Universidad Andres Bello, Santiago, Chile.
| |
Collapse
|
5
|
Endocytic Transport of Polyplex and Lipoplex siRNA Vectors in HeLa Cells. Pharm Res 2016; 33:2999-3011. [DOI: 10.1007/s11095-016-2022-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 08/12/2016] [Indexed: 12/11/2022]
|
6
|
Rewatkar PV, Sester DP, Parekh HS, Parat MO. Express in Vitro Plasmid Transfection Achieved with 16 + Asymmetric Peptide Dendrimers. ACS Biomater Sci Eng 2016; 2:438-445. [PMID: 33429545 DOI: 10.1021/acsbiomaterials.6b00033] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Asymmetric cationic amino acid-based dendrimers are highly branched chemically derived gene vectors developed to transport cargo such as plasmid DNA across the plasma membrane. We have previously demonstrated their propensity to enter cells that form caveolae, driven by positive charge density and promoted by arginine head groups. Caveolae are plasma membrane subdomains serving a number of cellular functions including endocytosis. Their formation requires membrane proteins (caveolins) and cytoplasmic proteins (cavins), so that gene disruption of either caveolin-1 or cavin-1 (also known as PTRF, i.e., polymerase I and transcript release factor) results in caveola deficiency. Here we evaluated the ability of a 16+ charged asymmetric arginine dendrimer to transfect plasmid DNA into cultured cells. We unveiled efficient transfection efficiencies (≥30%) 24-48 h after exposing the cells to dendrimer/pDNA complexes for only 5 min. Using wild type (WT) and caveolin-1 or PTRF gene-disrupted, i.e., caveola-deficient mouse embryo fibroblasts, we further show that caveolae promote pDNA transfection by 16+ charged asymmetric arginine dendrimers.
Collapse
Affiliation(s)
- Prarthana V Rewatkar
- School of Pharmacy, The University of Queensland, 20 Cornwall Street, Woolloongabba, Queensland 4102, Australia
| | - David P Sester
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Harendra S Parekh
- School of Pharmacy, The University of Queensland, 20 Cornwall Street, Woolloongabba, Queensland 4102, Australia
| | - Marie-Odile Parat
- School of Pharmacy, The University of Queensland, 20 Cornwall Street, Woolloongabba, Queensland 4102, Australia
| |
Collapse
|
7
|
Rewatkar PV, Parton RG, Parekh HS, Parat MO. Are caveolae a cellular entry route for non-viral therapeutic delivery systems? Adv Drug Deliv Rev 2015; 91:92-108. [PMID: 25579057 DOI: 10.1016/j.addr.2015.01.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 12/23/2014] [Accepted: 01/02/2015] [Indexed: 12/20/2022]
Abstract
The development of novel therapies increasingly relies on sophisticated delivery systems that allow the drug or gene expression-modifying agent of interest entry into cells. These systems can promote cellular targeting and/or entry, and they vary in size, charge, and functional group chemistry. Their optimization requires an in depth knowledge of the cellular routes of entry in normal and pathological states. Caveolae are plasma membrane invaginations that have the potential to undergo endocytosis. We critically review the literature exploring whether drug or nucleic acid delivery systems exploit and/or promote cellular entry via caveolae. A vast majority of studies employ pharmacological tools, co-localization experiments and very few make use of molecular tools. We provide clarification on how results of such studies should be interpreted and make suggestions for future studies.
Collapse
Affiliation(s)
- Prarthana V Rewatkar
- The University of Queensland, School of Pharmacy, 20 Cornwall Street, Woolloongabba, QLD 4102, Australia
| | - Robert G Parton
- The University of Queensland, Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, QLD 4072 Australia.
| | - Harendra S Parekh
- The University of Queensland, School of Pharmacy, 20 Cornwall Street, Woolloongabba, QLD 4102, Australia.
| | - Marie-Odile Parat
- The University of Queensland, School of Pharmacy, 20 Cornwall Street, Woolloongabba, QLD 4102, Australia.
| |
Collapse
|
8
|
Rodriguez L, Vallecorsa P, Battah S, Di Venosa G, Calvo G, Mamone L, Sáenz D, Gonzalez MC, Batlle A, MacRobert AJ, Casas A. Aminolevulinic acid dendrimers in photodynamic treatment of cancer and atheromatous disease. Photochem Photobiol Sci 2015; 14:1617-27. [DOI: 10.1039/c5pp00126a] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
ALA dendrimers are taken up by caveolae-mediated endocytosis in macrophages. Intracellular ALA release gives rise to PpIX synthesis and subsequent photosensitization of key cells in atheromas and tumour diseases.
Collapse
Affiliation(s)
- L. Rodriguez
- Centro de Investigaciones sobre Porfirinas y Porfirias (CIPYP). CONICET-Htal de Clínicas Gral. José de San Martín
- Ciudad de Buenos Aires
- Argentina
| | - P. Vallecorsa
- Centro de Investigaciones sobre Porfirinas y Porfirias (CIPYP). CONICET-Htal de Clínicas Gral. José de San Martín
- Ciudad de Buenos Aires
- Argentina
| | - S. Battah
- School of Biological Sciences
- University of Essex
- Wivenhoe Park CO4 3SQ
- UK
- Division of Surgery and Interventional Sciences and UCL Institute of Biomedical Engineering
| | - G. Di Venosa
- Centro de Investigaciones sobre Porfirinas y Porfirias (CIPYP). CONICET-Htal de Clínicas Gral. José de San Martín
- Ciudad de Buenos Aires
- Argentina
| | - G. Calvo
- Centro de Investigaciones sobre Porfirinas y Porfirias (CIPYP). CONICET-Htal de Clínicas Gral. José de San Martín
- Ciudad de Buenos Aires
- Argentina
| | - L. Mamone
- Centro de Investigaciones sobre Porfirinas y Porfirias (CIPYP). CONICET-Htal de Clínicas Gral. José de San Martín
- Ciudad de Buenos Aires
- Argentina
| | - D. Sáenz
- Centro de Investigaciones sobre Porfirinas y Porfirias (CIPYP). CONICET-Htal de Clínicas Gral. José de San Martín
- Ciudad de Buenos Aires
- Argentina
| | - M. C. Gonzalez
- Facultad de Ciencias Médicas
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP)
- CONICET-UNLP
- Argentina
| | - A. Batlle
- Centro de Investigaciones sobre Porfirinas y Porfirias (CIPYP). CONICET-Htal de Clínicas Gral. José de San Martín
- Ciudad de Buenos Aires
- Argentina
| | - A. J. MacRobert
- Division of Surgery and Interventional Sciences and UCL Institute of Biomedical Engineering
- University College London
- London W1W 7EJ
- UK
| | - A. Casas
- Centro de Investigaciones sobre Porfirinas y Porfirias (CIPYP). CONICET-Htal de Clínicas Gral. José de San Martín
- Ciudad de Buenos Aires
- Argentina
| |
Collapse
|
9
|
de la Fuente M, Jones MC, Santander-Ortega MJ, Mirenska A, Marimuthu P, Uchegbu I, Schätzlein A. A nano-enabled cancer-specific ITCH RNAi chemotherapy booster for pancreatic cancer. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 11:369-77. [PMID: 25267700 DOI: 10.1016/j.nano.2014.09.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 09/01/2014] [Accepted: 09/18/2014] [Indexed: 02/06/2023]
Abstract
UNLABELLED Gemcitabine is currently the standard therapy for pancreatic cancer. However, growing concerns over gemcitabine resistance mean that new combinatory therapies are required to prevent loss of efficacy with prolonged treatment. Here, we suggest that this could be achieved through co-administration of RNA interference agents targeting the ubiquitin ligase ITCH. Stable anti-ITCH siRNA and shRNA dendriplexes with a desirable safety profile were prepared using generation 3 poly(propylenimine) dendrimers (DAB-Am16). The complexes were efficiently taken up by human pancreatic cancer cells and produced a 40-60% decrease in ITCH RNA and protein expression in vitro (si/shRNA) and in a xenograft model of pancreatic cancer (shRNA). When co-administered with gemcitabine (100 mg/kg/week) at a subtherapeutic dose, treatment with ITCH-shRNA (3x 50 mg/week) was able to fully suppress tumour growth for 17 days, suggesting that downregulation of ITCH mediated by DAB-Am16/shRNA sensitizes pancreatic cancer to gemcitabine in an efficient and specific manner. FROM THE CLINICAL EDITOR Gemcitabine delivery to pancreatic cancer often results in the common problem of drug resistance. This team overcame the problem through co-administration of siRNA and shRNA dendriplexes targeting the ubiquitin ligase ITCH.
Collapse
Affiliation(s)
| | | | | | - Anja Mirenska
- UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX
| | | | - Ijeoma Uchegbu
- UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX
| | | |
Collapse
|
10
|
New views and insights into intracellular trafficking of drug-delivery systems by fluorescence fluctuation spectroscopy. Ther Deliv 2014; 5:173-88. [DOI: 10.4155/tde.13.148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Biomaterials in the nanometer size range can be engineered for site-specific delivery of drugs after injection into the blood circulation. However, translation of such nanomedicines from the bench to the bedside is still hindered by many extracellular and intracellular barriers. To realize the concept of targeted drug delivery with nanomedicines, research groups are studying intensively the extra- and intra-cellular mechanisms involved as a response to the physicochemical properties of the nanomedicines. In this review, we highlight the contributions of fluorescence fluctuations spectroscopy techniques to better understand, and in turn to bypass, the major hurdles to therapeutic delivery, focusing mostly on the intracellular dynamics of drug-delivery systems.
Collapse
|
11
|
Fichter KM, Ingle NP, McLendon PM, Reineke TM. Polymeric nucleic acid vehicles exploit active interorganelle trafficking mechanisms. ACS NANO 2013; 7:347-64. [PMID: 23234474 PMCID: PMC3586558 DOI: 10.1021/nn304218q] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Materials that self-assemble with nucleic acids into nanocomplexes (e.g. polyplexes) are widely used in many fundamental biological and biomedical experiments. However, understanding the intracellular transport mechanisms of these vehicles remains a major hurdle in their effective usage. Here, we investigate two polycation models, Glycofect (which slowly degrades via hydrolysis) and linear polyethyleneimine (PEI) (which does not rapidly hydrolyze), to determine the impact of polymeric structure on intracellular trafficking. Cells transfected using Glycofect underwent increasing transgene expression over the course of 40 h and remained benign over the course of 7 days. Transgene expression in cells transfected with PEI peaked at 16 h post-transfection and resulted in less than 10% survival after 7 days. While saccharide-containing Glycofect has a higher buffering capacity than PEI, polyplexes created with Glycofect demonstrate more sustained endosomal release, possibly suggesting an additional or alternative delivery mechanism to the classical "proton sponge mechanism". PEI appeared to promote release of DNA from acidic organelles more than Glycofect. Immunofluorescence images indicate that both Glycofect and linear PEI traffic oligodeoxynucleotides to the Golgi and endoplasmic reticulum, which may be a route towards nuclear delivery. However, Glycofect polyplexes demonstrated higher co-localization with the ER than PEI polyplexes, and co-localization experiments indicate the retrograde transport of polyplexes via COP I vesicles from the Golgi to the ER. We conclude that slow release and unique trafficking behaviors of Glycofect polyplexes may be due to the presence of saccharide units and the degradable nature of the polymer, allowing more efficacious and benign delivery.
Collapse
Affiliation(s)
- Katye M. Fichter
- Department Chemistry, Missouri State University, Springfield, MO
| | - Nilesh. P. Ingle
- Department of Chemistry, University of Minnesota-Twin Cities, Minneapolis, MN
| | - Patrick M. McLendon
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Theresa M. Reineke
- Department of Chemistry, University of Minnesota-Twin Cities, Minneapolis, MN
- Corresponding Author. Correspondence should be addressed to Professor Theresa M. Reineke, Department of Chemistry, University of Minnesota-Twin Cities, Minneapolis, MN. Phone: 612-624-8042.
| |
Collapse
|
12
|
Sajomsang W, Gonil P, Ruktanonchai UR, Petchsangsai M, Opanasopit P, Puttipipatkhachorn S. Effects of molecular weight and pyridinium moiety on water-soluble chitosan derivatives for mediated gene delivery. Carbohydr Polym 2012; 91:508-17. [PMID: 23121939 DOI: 10.1016/j.carbpol.2012.08.053] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 08/16/2012] [Accepted: 08/16/2012] [Indexed: 11/17/2022]
Abstract
The aim of this study is to investigate the effects of molecular weight, the pyridinium/trimethyl ammonium (Py/Tr) ratio, the nitrogen atoms (N) in the methylated N-(3-pyridylmethyl) chitosan chloride (M3-PyMeChC)/the phosphorus atoms (P) in DNA (N/P) ratio, and the physicochemical properties of nanopolyplexes on transfection efficiency. The water-soluble chitosan derivative, M3-PyMeChC, was used as a non-viral vector to deliver pEGFP-C2 into human hepatoma (Huh7) cell lines. The results revealed that higher molecular weight M3-PyMeChC was able to form complexes completely with DNA at lower N/P ratios than that with lower molecular weights, which led to higher transfection efficiency. Moreover, the M3-PyMeChC with higher Py/Tr ratios showed superior transfection efficiency at lower Py/Tr ratios at all N/P ratios studied. The highest transfection efficiency for the nanopolyplexes occurred for a molecular weight of 82kDa at a N/P ratio of 5. The results indicated that the hydrophobic effect of pyridinium moiety could enhance gene transfection efficiency, which can be attributed to the dissociation of DNA from nanopolyplexes. High Py/Tr ratios in nanopolyplexes tended to decrease cytotoxicity due to delocalization of positive charge into a pyridine ring while high N/P ratios and molecular weight increased cytotoxicity. Our results showed that the vector was able to spread the positive charge by delocalizing it into a heterocyclic ring, suggesting to a promising approach to mediate higher levels of gene transfection.
Collapse
Affiliation(s)
- Warayuth Sajomsang
- National Nanotechnology Center, National Science and Technology Development Agency, Pathumthani, Thailand.
| | | | | | | | | | | |
Collapse
|
13
|
Cellular uptake and intracellular trafficking of PEG-b-PLA polymeric micelles. Biomaterials 2012; 33:7233-40. [PMID: 22795850 DOI: 10.1016/j.biomaterials.2012.06.045] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 06/22/2012] [Indexed: 01/23/2023]
Abstract
Besides as an inert carrier for hydrophobic anticancer agents, polymeric micelles composed of di-block copolymer poly(ethylene glycol)-poly(lactic acid) (PEG-b-PLA) function as biological response modifiers including reversal of multidrug resistance in cancer. However, the uptake mechanisms and the subsequent intracellular trafficking remain to be elucidated. In this paper, we found that the uptake of PEG-b-PLA polymeric micelles incorporating nile red (M-NR) was significantly inhibited by both dynamin inhibitor dynasore and dynamin-2 dominant negative mutant (dynamin-2 K44A). Exogenously expressed caveolin-1 colocalized with M-NR and upregulated M-NR internalization in HepG2 cells expressing low level of endogenous caveolin-1, while caveolin-1 dominant negative mutant (caveolin-1 Y14F) significantly downregulated M-NR internalization in C6 cells expressing high level of endogenous caveolin-1. Exogenously expressed clathrin light chain A (clathrin LCa) did not mainly colocalize with the internalized M-NR and had no effect on M-NR uptake. These results suggested that dynamin- and caveolin-dependent but clathrin-independent endocytosis was involved in M-NR cellular uptake. We further found that M-NR colocalized with lysosome and microtubulin after internalization.
Collapse
|
14
|
On the cellular processing of non-viral nanomedicines for nucleic acid delivery: Mechanisms and methods. J Control Release 2012; 161:566-81. [DOI: 10.1016/j.jconrel.2012.05.020] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 05/11/2012] [Accepted: 05/11/2012] [Indexed: 11/24/2022]
|
15
|
Affiliation(s)
- Quanming Lin
- a College of Materials and Textiles, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Guohua Jiang
- b College of Materials and Textiles, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China, Key Laboratory of Advanced Textile Materials and Manufacturing Technology (ATMT), Zhejiang Sci-Tech University, Ministry of Education, Hangzhou 310018, P. R. China;,
| | - Kangkang Tong
- c College of Materials and Textiles, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| |
Collapse
|
16
|
Liu X, Liu C, Laurini E, Posocco P, Pricl S, Qu F, Rocchi P, Peng L. Efficient delivery of sticky siRNA and potent gene silencing in a prostate cancer model using a generation 5 triethanolamine-core PAMAM dendrimer. Mol Pharm 2012; 9:470-81. [PMID: 22208617 DOI: 10.1021/mp2006104] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Successful achievement of RNA interference in therapeutic applications requires safe and efficient vectors for siRNA delivery. In the present study, we demonstrate that a triethanolamine (TEA)-core PAMAM dendrimer of generation 5 (G(5)) is able to deliver sticky siRNAs bearing complementary A(n)/T(n) 3'-overhangs effectively to a prostate cancer model in vitro and in vivo and produce potent gene silencing of the heat shock protein 27, leading to a notable anticancer effect. The complementary A(n)/T(n) (n = 5 or 7) overhangs characteristic of these sticky siRNA molecules help the siRNA molecules self-assemble into "gene-like" longer double-stranded RNAs thus endowing a low generation dendrimer such as G(5) with greater delivery capacity. In addition, the A(n)/T(n) (n = 5 or 7) overhangs act as protruding molecular arms that allow the siRNA molecule to enwrap the dendrimer and promote a better interaction and stronger binding, ultimately contributing toward the improved delivery activity of G(5). Consequently, the low generation dendrimer G(5) in combination with sticky siRNA therapeutics may constitute a promising gene silencing-based approach for combating castration-resistant prostate tumors or other cancers and diseases, for which no effective treatment currently exists.
Collapse
Affiliation(s)
- Xiaoxuan Liu
- Département de Chimie, Centre Interdisciplinaire de Nanoscience de Marseille, CNRS UMR 7325, Aix-Marseille Université, 163 avenue de Luminy, 13288 Marseille, France
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Liu X, Wu J, Yammine M, Zhou J, Posocco P, Viel S, Liu C, Ziarelli F, Fermeglia M, Pricl S, Victorero G, Nguyen C, Erbacher P, Behr JP, Peng L. Structurally Flexible Triethanolamine Core PAMAM Dendrimers Are Effective Nanovectors for DNA Transfection in Vitro and in Vivo to the Mouse Thymus. Bioconjug Chem 2011; 22:2461-73. [DOI: 10.1021/bc200275g] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Xiaoxuan Liu
- Aix-Marseille
Université,
Centre Interdisciplinaire de Nanoscience de Marseille, CNRS UPR 3118, Département de Chimie, 163 avenue
de Luminy, 13288 Marseille cedex 09, France
- State Key Laboratory of Virology,
College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
| | - Jiangyu Wu
- Aix-Marseille
Université,
Centre Interdisciplinaire de Nanoscience de Marseille, CNRS UPR 3118, Département de Chimie, 163 avenue
de Luminy, 13288 Marseille cedex 09, France
- State Key Laboratory of Virology,
College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
| | - Miriam Yammine
- INSERM U928, 163 avenue de Luminy, 13288
Marseille cedex 09, France
| | - Jiehua Zhou
- State Key Laboratory of Virology,
College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
| | - Paola Posocco
- Molecular Simulation
Engineering
(MOSE) Laboratory, Department of Chemical Engineering, University of Trieste, Piazzale Europa 1, 34127 Trieste,
Italy
| | - Stephane Viel
- Aix-Marseille Université, LCP UMR 6264, Campus de Saint Jérôme,
av. Escadrille Normandie Niémen, case 512, 13013 Marseille,
France
| | - Cheng Liu
- State Key Laboratory of Virology,
College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
| | - Fabio Ziarelli
- Aix-Marseille Université, Fédération des Sciences Chimiques, Spectropole,
av. Escadrille Normandie Niémen, case 511, 13013 Marseille,
France
| | - Maurizio Fermeglia
- Molecular Simulation
Engineering
(MOSE) Laboratory, Department of Chemical Engineering, University of Trieste, Piazzale Europa 1, 34127 Trieste,
Italy
| | - Sabrina Pricl
- Molecular Simulation
Engineering
(MOSE) Laboratory, Department of Chemical Engineering, University of Trieste, Piazzale Europa 1, 34127 Trieste,
Italy
| | | | - Catherine Nguyen
- INSERM U928, 163 avenue de Luminy, 13288
Marseille cedex 09, France
| | - Patrick Erbacher
- Polyplus-transfection SA, Bioparc, Boulevard S. Brandt, BP90018, 67401 Illkirch,
France
| | - Jean-Paul Behr
- Laboratoire de Chimie Génétique,
Faculté de Pharmacie, CNRS UMR7514, 67401 Illkirch, France
| | - Ling Peng
- Aix-Marseille
Université,
Centre Interdisciplinaire de Nanoscience de Marseille, CNRS UPR 3118, Département de Chimie, 163 avenue
de Luminy, 13288 Marseille cedex 09, France
| |
Collapse
|
18
|
Perez AP, Cosaka ML, Romero EL, Morilla MJ. Uptake and intracellular traffic of siRNA dendriplexes in glioblastoma cells and macrophages. Int J Nanomedicine 2011; 6:2715-28. [PMID: 22114502 PMCID: PMC3218585 DOI: 10.2147/ijn.s25235] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Gene silencing using small interfering RNA (siRNA) is a promising new therapeutic approach for glioblastoma. The endocytic uptake and delivery of siRNA to intracellular compartments could be enhanced by complexation with polyamidoamine dendrimers. In the present work, the uptake mechanisms and intracellular traffic of siRNA/generation 7 dendrimer complexes (siRNA dendriplexes) were screened in T98G glioblastoma and J774 macrophages. METHODS The effect of a set of chemical inhibitors of endocytosis on the uptake and silencing capacity of dendriplexes was determined by flow cytometry. Colocalization of fluorescent dendriplexes with endocytic markers and occurrence of intracellular dissociation were assessed by confocal laser scanning microscopy. RESULTS Uptake of siRNA dendriplexes by T98G cells was reduced by methyl-β-cyclodextrin, and genistein, and cytochalasine D, silencing activity was reduced by genistein; dendriplexes colocalized with cholera toxin subunit B. Therefore, caveolin-dependent endocytosis was involved both in the uptake and silencing activity of siRNA dendriplexes. On the other hand, uptake of siRNA dendriplexes by J774 cells was reduced by methyl-β-cyclodextrin, genistein, chlorpromazine, chloroquine, cytochalasine D, and nocodazole, the silencing activity was not affected by chlorpromazine, genistein or chloroquine, and dendriplexes colocalized with transferrin and cholera toxin subunit B. Thus, both clathrin-dependent and caveolin-dependent endocytosis mediated the uptake and silencing activity of the siRNA dendriplexes. SiRNA dendriplexes were internalized at higher rates by T98G but induced lower silencing than in J774 cells. SiRNA dendriplexes showed relatively slow dissociation kinetics, and their escape towards the cytosol was not mediated by acidification independently of the uptake pathway. CONCLUSION The extent of cellular uptake of siRNA dendriplexes was inversely related to their silencing activity. The higher silencing activity of siRNA dendriplexes in J774 cells could be ascribed to the contribution of clathrin-dependent and caveolin-dependent endocytosis vs only caveolin-dependent endocytosis in T98G cells.
Collapse
Affiliation(s)
- Ana Paula Perez
- Programa de Nanomedicinas, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina
| | - Maria Luz Cosaka
- Programa de Nanomedicinas, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina
| | - Eder Lilia Romero
- Programa de Nanomedicinas, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina
| | - Maria Jose Morilla
- Programa de Nanomedicinas, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina
| |
Collapse
|
19
|
Abstract
Integrins have become key targets for molecular imaging and for selective delivery of anti-cancer agents. Here we review recent work concerning the targeted delivery of antisense and siRNA oligonucleotides via integrins. A variety of approaches have been used to link oligonucleotides to ligands capable of binding integrins with high specificity and affinity. This includes direct chemical conjugation, incorporating oligonucleotides into lipoplexes, and use of various polymeric nanocarriers including dendrimers. The ligand-oligonucleotide conjugate or complex associates selectively with the integrin, followed by internalization into endosomes and trafficking through subcellular compartments. Escape of antisense or siRNA from the endosome to the cytosol and nucleus may come about through endogenous trafficking mechanisms, or because of membrane disrupting capabilities built into the conjugate or complex. Thus a variety of useful strategies are available for using integrins to enhance the pharmacological efficacy of therapeutic oligonucleotides.
Collapse
|
20
|
Sunoqrot S, Bae JW, Jin SE, M Pearson R, Liu Y, Hong S. Kinetically controlled cellular interactions of polymer-polymer and polymer-liposome nanohybrid systems. Bioconjug Chem 2011; 22:466-74. [PMID: 21344902 DOI: 10.1021/bc100484t] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Although bioactive polymers such as cationic polymers have demonstrated potential as drug carriers and nonviral gene delivery vectors, high toxicity and uncontrolled, instantaneous cellular interactions of those vectors have hindered the successful implementation In Vivo. Fine control over the cellular interactions of a potential drug/gene delivery vector would be thus desirable. Herein, we have designed nanohybrid systems (100-150 nm in diameter) that combine the polycations with protective outer layers consisting of biodegradable polymeric nanoparticles (NPs) or liposomes. A commonly used polycation polyethylenimine (PEI) was employed after conjugation with rhodamine (RITC). The PEI-RITC conjugates were then encapsulated into (i) polymeric NPs made of either poly(lactide-co-glycolide) (PLGA) or poly(ethylene glycol)-b-poly(lactide-co-glycolide) (PEG-PLGA); or (ii) PEGylated liposomes, resulting in three nanohybrid systems. Through the nanohybridization, both cellular uptake and cytotoxicity of the nanohybrids were kinetically controlled. The cytotoxicity assay using MCF-7 cells revealed that liposome-based nanohybrids exhibited the least toxicity, followed by PEG-PLGA- and PLGA-based NPs after 24 h incubation. The different kinetics of cellular uptake was also observed, the liposome-based systems being the fastest and PLGA-based systems being the slowest. The results present a potential delivery platform with enhanced control over its biological interaction kinetics and passive targeting capability through size control.
Collapse
Affiliation(s)
- Suhair Sunoqrot
- Departments of †Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | | | | | | | | | | |
Collapse
|
21
|
Abstract
The impact that nanotechnology may have on life and medical sciences is immense and includes novel therapies as much as novel diagnostic and imaging tools, often offering the possibility to combine the two. It is, therefore, of the essence to understand and control the interactions that nanomaterials can have with cells, first at an individual level, focusing on, e.g., binding and internalization events, and then at a tissue level, where diffusion and long-range transport add further complications. Here, we present experimental methods based on selective labeling techniques and the use of effectors for a qualitative and quantitative evaluation of endocytic phenomena involving nanoparticles. The understanding of the cell-material interactions arising from these tests can then form the basis for a model-based evaluation of nanoparticles behavior in 3D tissues.
Collapse
|
22
|
Zaki NM, Tirelli N. Gateways for the intracellular access of nanocarriers: a review of receptor-mediated endocytosis mechanisms and of strategies in receptor targeting. Expert Opin Drug Deliv 2010; 7:895-913. [PMID: 20629604 DOI: 10.1517/17425247.2010.501792] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
IMPORTANCE OF THE FIELD The last 10 years have seen a dramatic growth in understanding and controlling how complex, drug-loaded (nano)structures, as well as pathogens, or biopharmaceuticals can gather access to the cytoplasm, which is a key step to increasing the effectiveness of their action. AREAS COVERED IN THIS REVIEW The review offers an updated overview of the current knowledge of endocytic processes; furthermore, the cell surface receptors most commonly used in drug delivery are here discussed on the basis of their reported internalization mechanisms, with examples of their use as nanocarrier targets taken from the most recent scientific literature. WHAT THE READER WILL GAIN Knowledge of molecular biology details is increasingly necessary for a rational design of drug delivery systems. Here, the aim is to provide the reader with an attempt to link a mechanistic knowledge of endocytic mechanisms with the identification of appropriate targets (internalization receptors) for nanocarriers. TAKE HOME MESSAGE Much advance is still needed to create a complete and coherent biological picture of endocytosis, but current knowledge already allows individuation of a good number of targetable groups for a predetermined intracellular fate of nanocarriers.
Collapse
Affiliation(s)
- Noha M Zaki
- Ain Shams University, Department of Pharmaceutics, Faculty of Pharmacy, Monazamet El Wehda El Afrikia St, El Abbassia, Cairo, Egypt
| | | |
Collapse
|
23
|
Beebe SJ, Schoenbach KH, Heller R. Bioelectric applications for treatment of melanoma. Cancers (Basel) 2010; 2:1731-70. [PMID: 24281185 PMCID: PMC3837335 DOI: 10.3390/cancers2031731] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Revised: 09/14/2010] [Accepted: 09/15/2010] [Indexed: 01/04/2023] Open
Abstract
Two new cancer therapies apply bioelectric principles. These methods target tumor structures locally and function by applying millisecond electric fields to deliver plasmid DNA encoding cytokines using electrogene transfer (EGT) or by applying rapid rise-time nanosecond pulsed electric fields (nsPEFs). EGT has been used to locally deliver cytokines such as IL-12 to activate an immune response, resulting in bystander effects. NsPEFs locally induce apoptosis-like effects and affect vascular networks, both promoting tumor demise and restoration of normal vascular homeostasis. EGT with IL-12 is in melanoma clinical trials and nsPEFs are used in models with B16F10 melanoma in vitro and in mice. Applications of bioelectrics, using conventional electroporation and extensions of it, provide effective alternative therapies for melanoma.
Collapse
Affiliation(s)
- Stephen J Beebe
- Frank Reidy Research Center for Bioelectrics/Old Dominion University 4211 Monarch Way, Suite 300, Norfolk, Virginia 23508, USA.
| | | | | |
Collapse
|
24
|
Chemical vectors for gene delivery: uptake and intracellular trafficking. Curr Opin Biotechnol 2010; 21:640-5. [PMID: 20674331 DOI: 10.1016/j.copbio.2010.07.003] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 07/06/2010] [Indexed: 01/07/2023]
Abstract
Chemical vectors for non-viral gene delivery are based on engineered DNA nanoparticles produced with various range of macromolecules suitable to mimic some viral functions required for gene transfer. Many efforts have been undertaken these past years to identify cellular barriers that have to be passed for this issue. Here, we summarize the current status of knowledge on the uptake mechanism of DNA nanoparticles made with polymers and liposomes, their endosomal escape, cytosolic diffusion, and nuclear import of pDNA. Studies reported these past years regarding pDNA nanoparticles endocytosis indicated that there is no clear evident relationship between the ways of entry and the transfection efficiency. By contrast, the sequestration of pDNA in intracellular vesicles and the low number of pDNA close to the nuclear envelop are identified as the major intracellular barriers. So, intensive investigations to increase the cytosolic delivery of pDNA and its migration toward nuclear pores make sense to bring the transfection efficiency closer to that of viruses.
Collapse
|
25
|
Pavan GM, Albertazzi L, Danani A. Ability to adapt: different generations of PAMAM dendrimers show different behaviors in binding siRNA. J Phys Chem B 2010; 114:2667-75. [PMID: 20146540 DOI: 10.1021/jp100271w] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
This paper reports a molecular dynamic study to explore the diverse behavior of different generations of poly(amidoamine) (PAMAM) dendrimers in binding siRNA. Our models show good accordance with experimental measurements. Simulations demonstrate that the molecular flexibility of PAMAMs plays a crucial role in the binding event, which is controlled by the modulation between enthalpy and entropy of binding. Importantly, the ability of dendrimers to adapt to siRNA is strongly dependent on the generation and on the pH due to backfolding. While G4 demonstrates good adaptability to siRNA, G6 behaves like a rigid sphere with a consistent loss in the binding affinity. G5 shows a hybrid behavior, maintaining rigid and flexible aspects, with a strong dependence of its properties on the pH. To define the "best binder", the mere energetic definition of binding affinity appears to be no longer effective and a novel concept of "efficiency" should be considered, being the balance between enthalpy and entropy of binding indivisible from the structural flexibility. With this aim, we propose an original criterion to define and rank the ability of these molecules to adapt their structure to bind a charged target.
Collapse
Affiliation(s)
- Giovanni M Pavan
- University for Applied Sciences of Southern Switzerland (SUPSI)-Institute of Computer Integrated Manufacturing for Sustainable Innovation, Centro Galleria 2, Manno, CH-6928, Switzerland.
| | | | | |
Collapse
|
26
|
Jain K, Kesharwani P, Gupta U, Jain NK. Dendrimer toxicity: Let's meet the challenge. Int J Pharm 2010; 394:122-42. [PMID: 20433913 DOI: 10.1016/j.ijpharm.2010.04.027] [Citation(s) in RCA: 477] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2010] [Revised: 04/19/2010] [Accepted: 04/19/2010] [Indexed: 12/15/2022]
Abstract
Dendrimers are well-defined, versatile polymeric architecture with properties resembling biomolecules. Dendritic polymers emerged as outstanding carrier in modern medicine system because of its derivatisable branched architecture and flexibility in modifying it in numerous ways. Dendritic scaffold has been found to be suitable carrier for a variety of drugs including anticancer, anti-viral, anti-bacterial, antitubercular etc., with capacity to improve solubility and bioavailability of poorly soluble drugs. In spite of extensive applicability in pharmaceutical field, the use of dendrimers in biological system is constrained because of inherent toxicity associated with them. This toxicity is attributed to the interaction of surface cationic charge of dendrimers with negatively charged biological membranes in vivo. Interaction of dendrimers with biological membranes results in membrane disruption via nanohole formation, membrane thinning and erosion. Dendrimer toxicity in biological system is generally characterized by hemolytic toxicity, cytotoxicity and hematological toxicity. To minimize this toxicity two strategies have been utilized; first, designing and synthesis of biocompatible dendrimers; and second, masking of peripheral charge of dendrimers by surface engineering. Biocompatible dendrimers can be synthesized by employing biodegradable core and branching units or utilizing intermediates of various metabolic pathways. Dendrimer biocompatibility has been evaluated in vitro and in vivo for efficient presentation of biological performance. Surface engineering masks the cationic charge of dendrimer surface either by neutralization of charge, for example PEGylation, acetylation, carbohydrate and peptide conjugation; or by introducing negative charge such as half generation dendrimers. Neutral and negatively charged dendrimers do not interact with biological environment and hence are compatible for clinical applications as elucidated by various studies examined in this review. Chemical modification of the surface is an important strategy to overcome the toxicity problems associated with the dendrimers. The present review emphasizes on the approaches available to overcome the cationic toxicity inherently associated with the dendrimers.
Collapse
Affiliation(s)
- Keerti Jain
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences, Dr. H. S. Gour University Sagar (M.P.) 470003 India
| | | | | | | |
Collapse
|
27
|
Albertazzi L, Serresi M, Albanese A, Beltram F. Dendrimer Internalization and Intracellular Trafficking in Living Cells. Mol Pharm 2010; 7:680-8. [DOI: 10.1021/mp9002464] [Citation(s) in RCA: 153] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Lorenzo Albertazzi
- NEST, Scuola Normale Superiore and Istituto Nanoscienze-CNR, I-56127 Pisa, Italy, and IIT@NEST, Center for Nanotechnology Innovation, I-56127 Pisa, Italy
| | - Michela Serresi
- NEST, Scuola Normale Superiore and Istituto Nanoscienze-CNR, I-56127 Pisa, Italy, and IIT@NEST, Center for Nanotechnology Innovation, I-56127 Pisa, Italy
| | - Alberto Albanese
- NEST, Scuola Normale Superiore and Istituto Nanoscienze-CNR, I-56127 Pisa, Italy, and IIT@NEST, Center for Nanotechnology Innovation, I-56127 Pisa, Italy
| | - Fabio Beltram
- NEST, Scuola Normale Superiore and Istituto Nanoscienze-CNR, I-56127 Pisa, Italy, and IIT@NEST, Center for Nanotechnology Innovation, I-56127 Pisa, Italy
| |
Collapse
|
28
|
|
29
|
Qi R, Mullen DG, Baker JR, Holl MMB. The mechanism of polyplex internalization into cells: testing the GM1/caveolin-1 lipid raft mediated endocytosis pathway. Mol Pharm 2010; 7:267-79. [PMID: 20025295 PMCID: PMC2826151 DOI: 10.1021/mp900241t] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The GM1/caveolin-1 lipid raft mediated endocytosis mechanism was explored for generation 5 and 7 poly(amidoamine) dendrimer polyplexes employing the Cos-7, 293A, C6, HeLa, KB, and HepG2 cell lines. Expression levels of GM1 and caveolin-1 were measured using dot blot and Western blot, respectively. The level of GM1 in the cell plasma membrane was adjusted by incubation with exogenous GM1 or ganglioside inhibitor PPMP, and the level of CAV-1 was adjusted by upregulation with the adenovirus vector expressed caveolin-1 (AdCav-1). Cholera toxin B subunit was employed as a positive control for uptake in all cases. No evidence was found for a GM1/caveolin-1 lipid raft mediated endocytosis mechanism for the generation 5 and 7 poly(amidoamine) dendrimer polyplexes.
Collapse
Affiliation(s)
- Rong Qi
- Peking University Institute of Cardiovascular Sciences, Peking University Health Science Center, Peking University, Beijing 100083, China
| | | | | | | |
Collapse
|
30
|
Hong S, Rattan R, Majoros IJ, Mullen DG, Peters JL, Shi X, Bielinska AU, Blanco L, Orr BG, Baker JR, Holl MMB. The role of ganglioside GM1 in cellular internalization mechanisms of poly(amidoamine) dendrimers. Bioconjug Chem 2009; 20:1503-13. [PMID: 19583240 DOI: 10.1021/bc900029k] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Generation 7 (G7) poly(amidoamine) (PAMAM) dendrimers with amine, acetamide, and carboxylate end groups were prepared to investigate polymer/cell membrane interactions in vitro. G7 PAMAM dendrimers were used in this study because higher-generation of dendrimers are more effective in permeabilization of cell plasma membranes and in the formation of nanoscale holes in supported lipid bilayers than smaller, lower-generation dendrimers. Dendrimer-based conjugates were characterized by (1)H NMR, UV/vis spectroscopy, GPC, HPLC, and CE. Positively charged amine-terminated G7 dendrimers (G7-NH(2)) were observed to internalize into KB, Rat2, and C6 cells at a 200 nM concentration. By way of contrast, neither negatively charged G7 carboxylate-terminated dendrimers (G7-COOH) nor neutral acetamide-terminated G7 dendrimers (G7-Ac) associated with the cell plasma membrane or internalized under similar conditions. A series of in vitro experiments employing endocytic markers cholera toxin subunit B (CTB), transferrin, and GM(1)-pyrene were performed to further investigate mechanisms of dendrimer internalization into cells. G7-NH(2) dendrimers colocalized with CTB; however, experiments with C6 cells indicated that internalization of G7-NH(2) was not ganglioside GM(1) dependent. The G7/CTB colocalization was thus ascribed to an artifact of direct interaction between the two species. The presence of GM(1) in the membrane also had no effect upon XTT assays of cell viability or lactate dehydrogenase (LDH) assays of membrane permeability.
Collapse
Affiliation(s)
- Seungpyo Hong
- Program in Macromolecular Science and Engineering and Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Theoharis S, Krueger U, Tan PH, Haskard DO, Weber M, George AJ. Targeting gene delivery to activated vascular endothelium using anti E/P-Selectin antibody linked to PAMAM dendrimers. J Immunol Methods 2009; 343:79-90. [DOI: 10.1016/j.jim.2008.12.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2008] [Revised: 11/26/2008] [Accepted: 12/17/2008] [Indexed: 02/08/2023]
|
32
|
Saovapakhiran A, D’Emanuele A, Attwood D, Penny J. Surface Modification of PAMAM Dendrimers Modulates the Mechanism of Cellular Internalization. Bioconjug Chem 2009; 20:693-701. [DOI: 10.1021/bc8002343] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Angkana Saovapakhiran
- School of Pharmacy and Pharmaceutical Sciences, The University of Manchester, Oxford Road, Manchester M13 9PT, and School of Pharmacy and Pharmaceutical Sciences, University of Central Lancashire, Preston, Lancashire PR1 2HE, United Kingdom
| | - Antony D’Emanuele
- School of Pharmacy and Pharmaceutical Sciences, The University of Manchester, Oxford Road, Manchester M13 9PT, and School of Pharmacy and Pharmaceutical Sciences, University of Central Lancashire, Preston, Lancashire PR1 2HE, United Kingdom
| | - David Attwood
- School of Pharmacy and Pharmaceutical Sciences, The University of Manchester, Oxford Road, Manchester M13 9PT, and School of Pharmacy and Pharmaceutical Sciences, University of Central Lancashire, Preston, Lancashire PR1 2HE, United Kingdom
| | - Jeffrey Penny
- School of Pharmacy and Pharmaceutical Sciences, The University of Manchester, Oxford Road, Manchester M13 9PT, and School of Pharmacy and Pharmaceutical Sciences, University of Central Lancashire, Preston, Lancashire PR1 2HE, United Kingdom
| |
Collapse
|
33
|
|
34
|
Abstract
This article provides an overview of principles and barriers relevant to intracellular drug and gene transport, accumulation and retention (collectively called as drug delivery) by means of nanovehicles (NV). The aim is to deliver a cargo to a particular intracellular site, if possible, to exert a local action. Some of the principles discussed in this article apply to noncolloidal drugs that are not permeable to the plasma membrane or to the blood-brain barrier. NV are defined as a wide range of nanosized particles leading to colloidal objects which are capable of entering cells and tissues and delivering a cargo intracelullarly. Different localization and targeting means are discussed. Limited discussion on pharmacokinetics and pharmacodynamics is also presented. NVs are contrasted to micro-delivery and current nanotechnologies which are already in commercial use. Newer developments in NV technologies are outlined and future applications are stressed. We also briefly review the existing modeling tools and approaches to quantitatively describe the behavior of targeted NV within the vascular and tumor compartments, an area of particular importance. While we list "elementary" phenomena related to different level of complexity of delivery to cancer, we also stress importance of multi-scale modeling and bottom-up systems biology approach.
Collapse
Affiliation(s)
- Ales Prokop
- Department of Chemical Engineering, 24th Avenue & Garland Avenues, 107 Olin Hall, Vanderbilt University, Nashville, Tennessee 37235, USA.
| | | |
Collapse
|
35
|
Kelly CV, Leroueil PR, Nett EK, Wereszczynski JM, Baker JR, Orr BG, Banaszak Holl MM, Andricioaei I. Poly(amidoamine) dendrimers on lipid bilayers I: Free energy and conformation of binding. J Phys Chem B 2008; 112:9337-45. [PMID: 18620450 DOI: 10.1021/jp801377a] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Third-generation (G3) poly(amidoamine) (PAMAM) dendrimers are simulated approaching 1,2-dimyristoyl- sn-glycero-3-phosphocholine (DMPC) bilayers with fully atomistic molecular dynamics, which enables the calculation of a free energy profile along the approach coordinate. Three different dendrimer terminations are examined: protonated primary amine, uncharged acetamide, and deprotonated carboxylic acid. As the dendrimer and lipids become closer, their attractive force increases (up to 240 pN) and the dendrimer becomes deformed as it interacts with the lipids. The total energy release upon binding of a G3-NH3+, G3-Ac, or G3-COO- dendrimer to a DMPC bilayer is, respectively, 36, 26, or 47 kcal/mol or, equivalently, 5.2, 3.2, or 4.7x10(-3) kcal/g. These results are analyzed in terms of the dendrimers' size, shape, and atomic distributions as well as proximity of individual lipid molecules and particular lipid atoms to the dendrimer. For example, an area of 9.6, 8.2, or 7.9 nm2 is covered on the bilayer for the G3-NH3+, G3-Ac, or G3-COO- dendrimers, respectively, while interacting strongly with 18-13 individual lipid molecules.
Collapse
Affiliation(s)
- Christopher V Kelly
- Applied Physics Program, Biophysics, Graham Environmental Sustainability Institute, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Kelly CV, Leroueil PR, Orr BG, Banaszak Holl MM, Andricioaei I. Poly(amidoamine) dendrimers on lipid bilayers II: Effects of bilayer phase and dendrimer termination. J Phys Chem B 2008; 112:9346-53. [PMID: 18620451 DOI: 10.1021/jp8013783] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The molecular structures and enthalpy release of poly(amidoamine) (PAMAM) dendrimers binding to 1,2-dimyristoyl- sn-glycero-3-phosphocholine (DMPC) bilayers were explored through atomistic molecular dynamics. Three PAMAM dendrimer terminations were examined: protonated primary amine, neutral acetamide, and deprotonated carboxylic acid. Fluid and gel lipid phases were examined to extract the effects of lipid tail mobility on the binding of generation-3 dendrimers, which are directly relevant to the nanoparticle interactions involving lipid rafts, endocytosis, lipid removal, and/or membrane pores. Upon binding to gel phase lipids, dendrimers remained spherical, had a constant radius of gyration, and approximately one-quarter of the terminal groups were in close proximity to the lipids. In contrast, upon binding to fluid phase bilayers, dendrimers flattened out with a large increase in their asphericity and radii of gyration. Although over twice as many dendrimer-lipid contacts were formed on fluid versus gel phase lipids, the dendrimer-lipid interaction energy was only 20% stronger. The greatest enthalpy release upon binding was between the charged dendrimers and the lipid bilayer. However, the stronger binding to fluid versus gel phase lipids was driven by the hydrophobic interactions between the inner dendrimer and lipid tails.
Collapse
Affiliation(s)
- Christopher V Kelly
- Applied Physics Program, Biophysics, Graham Environmental Sustainability Institute, Department of Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | | | |
Collapse
|
37
|
Vicente J, Abad JA, López-Nicolás RM. Synthesis of molecular chains: phenylene thioether and sulfoxide oligomers. Tetrahedron 2008. [DOI: 10.1016/j.tet.2008.04.108] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
38
|
Germershaus O, Mao S, Sitterberg J, Bakowsky U, Kissel T. Gene delivery using chitosan, trimethyl chitosan or polyethylenglycol-graft-trimethyl chitosan block copolymers: Establishment of structure–activity relationships in vitro. J Control Release 2008; 125:145-54. [DOI: 10.1016/j.jconrel.2007.10.013] [Citation(s) in RCA: 169] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2007] [Revised: 10/10/2007] [Accepted: 10/15/2007] [Indexed: 01/18/2023]
|
39
|
How SE, Unciti-Broceta A, Sánchez-Martín RM, Bradley M. Solid-phase synthesis of a lysine-capped bis-dendron with remarkable DNA delivery abilities. Org Biomol Chem 2008; 6:2266-9. [DOI: 10.1039/b804771e] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
40
|
Abstract
Intracellular trafficking of membrane-coated vesicles represents a fundamental process that controls the architecture of different intracellular compartments and communication between the cell and its environment. Major trafficking pathways consist of an inward flux of endocytic vesicles from the plasma membrane and an outward flux of exocytic vesicles to the plasma membrane. This overview describes a number of molecular biology tools commonly used to analyze endocytic and exocytic pathways. The overall emphasis is on major proteins responsible for vesicle formation, recognition, and fusion. These include components of vesicle coats, adaptor complexes, SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) proteins, and Rab guanosine 5'-triphosphatases (GTPases), which represent attractive targets for genetic manipulation aimed at unraveling mechanisms of endocytosis and exocytosis.
Collapse
Affiliation(s)
- Elena V Vassilieva
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | | |
Collapse
|
41
|
Theoharis S, Manunta M, Tan PH. Gene delivery to vascular endothelium using chemical vectors: implications for cardiovascular gene therapy. Expert Opin Biol Ther 2007; 7:627-43. [PMID: 17477801 DOI: 10.1517/14712598.7.5.627] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The vascular endothelium is an attractive target for gene therapy because of its accessibility and its importance in the pathophysiology of a wide range of cardiovascular conditions. In general, viral methods have been shown to be very effective at delivering genes to endothelium. The immunogenicity and pathogenicity associated with viral vectors have led increased efforts to seek alternative means of 'ferrying' therapeutic genes to endothelium or to decrease the short-comings of viral vectors. This paper reviews developments in non-viral technology. In addition, discussion also covers the mechanisms whereby existing chemical vectors deliver DNA to cells. Understanding the pathways of vector internalisation and intracellular traffic is important in developing strategies to improve vector technology. The authors propose that the chemical vector may represent a robust and versatile technology to 'ferry' therapeutic genes to vascular endothelium in order to modify the endothelial dysfunction associated with many cardiovascular diseases.
Collapse
Affiliation(s)
- Stefanos Theoharis
- Imperial College London, Department of Immunology, Division of Medicine, Hammersmith Hospital, Du Cane Road, London W12 ONN, UK
| | | | | |
Collapse
|