Noncanonical self-assembly of multifunctional DNA nanoflowers for biomedical applications.

DNA nanotechnology has been extensively explored to assemble various functional nanostructures for versatile applications. Mediated by Watson-Crick base-pairing, these DNA nanostructures have been conventionally assembled through hybridization of many short DNA building blocks. Here we report the noncanonical self-assembly of multifunctional DNA nanostructures, termed as nanoflowers (NFs), and the versatile biomedical applications. These NFs were assembled from long DNA building blocks generated via rolling circle replication (RCR) of a designer template. NF assembly was driven by liquid crystallization and dense packaging of building blocks, without relying on Watson-Crick base-pairing between DNA strands, thereby avoiding the otherwise conventional complicated DNA sequence design. NF sizes were readily tunable in a wide range, by simply adjusting such parameters as assembly time and template sequences. NFs were exceptionally resistant to nuclease degradation, denaturation, or dissociation at extremely low concentration, presumably resulting from the dense DNA packaging in NFs. The exceptional biostability is critical for biomedical applications. By rational design, NFs can be readily incorporated with myriad functional moieties. All these properties make NFs promising for versatile applications. As a proof-of-principle demonstration, in this study, NFs were integrated with aptamers, bioimaging agents, and drug loading sites, and the resultant multifunctional NFs were demonstrated for selective cancer cell recognition, bioimaging, and targeted anticancer drug delivery.

[1]  T. Yasuda,et al.  Measurement of deoxyribonuclease I activity in human tissues and body fluids by a single radial enzyme-diffusion method. , 1993, Clinical chemistry.

[2]  R. Chari,et al.  Targeted cancer therapy: conferring specificity to cytotoxic drugs. , 2008, Accounts of chemical research.

[3]  Steve Goodison,et al.  A Novel Aptamer Developed for Breast Cancer Cell Internalization , 2012, ChemMedChem.

[4]  A. Fire,et al.  Rolling replication of short DNA circles. , 1995, Proceedings of the National Academy of Sciences of the United States of America.

[5]  Bingling Li,et al.  Real-time detection of isothermal amplification reactions with thermostable catalytic hairpin assembly. , 2013, Journal of the American Chemical Society.

[6]  D. Pyshnyi,et al.  Circulating DNA and DNase Activity in Human Blood , 2006, Annals of the New York Academy of Sciences.

[7]  Simon A McManus,et al.  Turning a kinase deoxyribozyme into a sensor. , 2013, Journal of the American Chemical Society.

[8]  S. Santra,et al.  Cell-specific, activatable, and theranostic prodrug for dual-targeted cancer imaging and therapy. , 2011, Journal of the American Chemical Society.

[9]  Hanadi F Sleiman,et al.  Rolling circle amplification-templated DNA nanotubes show increased stability and cell penetration ability. , 2012, Journal of the American Chemical Society.

[10]  D. Shangguan,et al.  Aptamers evolved from live cells as effective molecular probes for cancer study , 2006, Proceedings of the National Academy of Sciences.

[11]  G. Schettini,et al.  Antisense oligonucleotides as therapeutic agents. , 1997, Pharmacological research.

[12]  P. Brown,et al.  A DNA microarray system for analyzing complex DNA samples using two-color fluorescent probe hybridization. , 1996, Genome research.

[13]  Xiangling Xiong,et al.  Self-assembled aptamer-based drug carriers for bispecific cytotoxicity to cancer cells. , 2012, Chemistry, an Asian journal.

[14]  Weihong Tan,et al.  Self-assembled, aptamer-tethered DNA nanotrains for targeted transport of molecular drugs in cancer theranostics , 2013, Proceedings of the National Academy of Sciences.

[15]  F. Livolant,et al.  Cholesteric organization of DNA in the stallion sperm head. , 1984, Tissue & cell.

[16]  Hao Yan,et al.  Rolling-circle amplification of a DNA nanojunction. , 2006, Angewandte Chemie.

[17]  Weian Zhao,et al.  Bioinspired multivalent DNA network for capture and release of cells , 2012, Proceedings of the National Academy of Sciences.

[18]  G. Weiner,et al.  Immunostimulatory oligodeoxynucleotides containing the CpG motif are effective as immune adjuvants in tumor antigen immunization. , 1997, Proceedings of the National Academy of Sciences of the United States of America.

[19]  Anthony D. Keefe,et al.  Aptamers as therapeutics , 2010, Nature Reviews Drug Discovery.

[20]  F. Livolant,et al.  The highly concentrated liquid-crystalline phase of DNA is columnar hexagonal , 1989, Nature.

[21]  D. Segal,et al.  Evidence for rolling circle replication of tandem genes in Drosophila , 2005, Nucleic acids research.

[22]  Robert B. Hartlage,et al.  This PDF file includes: Materials and Methods , 2009 .

[23]  N. Clark,et al.  Liquid crystal self-assembly of random-sequence DNA oligomers , 2012, Proceedings of the National Academy of Sciences.

[24]  N. Seeman Nanomaterials based on DNA. , 2010, Annual review of biochemistry.

[25]  T. T. Herskovits Nonaqueous solutions of DNA; denaturation by urea and its methyl derivatives. , 1963, Biochemistry.

[26]  Jon P Anderson,et al.  Fluorescent structural DNA nanoballs functionalized with phosphate-linked nucleotide triphosphates. , 2007, Nano letters.

[27]  Hao Yan,et al.  Rolling circle enzymatic replication of a complex multi-crossover DNA nanostructure. , 2007, Journal of the American Chemical Society.

[28]  Weihong Tan,et al.  Cell-specific aptamer probes for membrane protein elucidation in cancer cells. , 2008, Journal of proteome research.

[29]  Dongyu Liu,et al.  Rolling Circle DNA Synthesis: Small Circular Oligonucleotides as Efficient Templates for DNA Polymerases. , 1996, Journal of the American Chemical Society.

[30]  Joseph M. DeSimone,et al.  Strategies in the design of nanoparticles for therapeutic applications , 2010, Nature Reviews Drug Discovery.

[31]  M. Van Ranst,et al.  Rolling-circle amplification of viral DNA genomes using phi29 polymerase. , 2009, Trends in microbiology.

[32]  J. Wong,et al.  Birefringence and DNA Condensation of Liquid Crystalline Chromosomes , 2010, Eukaryotic Cell.

[33]  Noel A. Clark,et al.  End-to-End Stacking and Liquid Crystal Condensation of 6– to 20–Base Pair DNA Duplexes , 2007, Science.

[34]  D. Luo,et al.  A mechanical metamaterial made from a DNA hydrogel. , 2012, Nature nanotechnology.

[35]  U. Landegren,et al.  In situ genotyping individual DNA molecules by target-primed rolling-circle amplification of padlock probes , 2004, Nature Methods.

[36]  Michael W. Davidson,et al.  Multiple liquid crystal phases of DNA at high concentrations , 1988, Nature.

[37]  Daniel K. Bonner,et al.  Self-assembled RNA interference microsponges for efficient siRNA delivery. , 2012, Nature materials.

[38]  Peixuan Guo,et al.  Thermodynamically Stable RNA three-way junctions as platform for constructing multi-functional nanoparticles for delivery of therapeutics , 2011, Nature Nanotechnology.

[39]  Chunhai Fan,et al.  Lab in a tube: ultrasensitive detection of microRNAs at the single-cell level and in breast cancer patients using quadratic isothermal amplification. , 2013, Journal of the American Chemical Society.

[40]  J. Barton,et al.  Mechanisms for DNA charge transport. , 2010, Chemical reviews.

[41]  J. Karp,et al.  Nanocarriers as an Emerging Platform for Cancer Therapy , 2022 .

[42]  Donald E. Olins,et al.  Chromatin history: our view from the bridge , 2003, Nature Reviews Molecular Cell Biology.

[43]  Timothy B. Stockwell,et al.  The Sequence of the Human Genome , 2001, Science.