Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 267
Filter
1.
Proc Natl Acad Sci U S A ; 121(11): e2307809121, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38437543

ABSTRACT

Rapid advances in nucleic acid therapies highlight the immense therapeutic potential of genetic therapeutics. Lipid nanoparticles (LNPs) are highly potent nonviral transfection agents that can encapsulate and deliver various nucleic acid therapeutics, including but not limited to messenger RNA (mRNA), silencing RNA (siRNA), and plasmid DNA (pDNA). However, a major challenge of targeted LNP-mediated systemic delivery is the nanoparticles' nonspecific uptake by the liver and the mononuclear phagocytic system, due partly to the adsorption of endogenous serum proteins onto LNP surfaces. Tunable LNP surface chemistries may enable efficacious delivery across a range of organs and cell types. Here, we describe a method to electrostatically adsorb bioactive polyelectrolytes onto LNPs to create layered LNPs (LLNPs). LNP cores varying in nucleic acid cargo and component lipids were stably layered with four biologically relevant polyanions: hyaluronate (HA), poly-L-aspartate (PLD), poly-L-glutamate (PLE), and polyacrylate (PAA). We further investigated the impact of the four surface polyanions on the transfection and uptake of mRNA- and pDNA-loaded LNPs in cell cultures. PLD- and PLE-LLNPs increased mRNA transfection twofold over unlayered LNPs in immune cells. HA-LLNPs increased pDNA transfection rates by more than twofold in epithelial and immune cells. In a healthy C57BL/6 murine model, PLE- and HA-LLNPs increased transfection by 1.8-fold to 2.5-fold over unlayered LNPs in the liver and spleen. These results suggest that LbL assembly is a generalizable, highly tunable platform to modify the targeting specificity, stability, and transfection efficacy of LNPs, as well as incorporate other charged targeting and therapeutic molecules into these systems.


Subject(s)
Liposomes , Nanoparticles , Animals , Mice , Polyelectrolytes , Adsorption , Static Electricity , Transfection , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Glutamic Acid
2.
Proc Natl Acad Sci U S A ; 121(11): e2307802121, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38437557

ABSTRACT

RNA interference (RNAi) therapeutics are an emerging class of medicines that selectively target mRNA transcripts to silence protein production and combat disease. Despite the recent progress, a generalizable approach for monitoring the efficacy of RNAi therapeutics without invasive biopsy remains a challenge. Here, we describe the development of a self-reporting, theranostic nanoparticle that delivers siRNA to silence a protein that drives cancer progression while also monitoring the functional activity of its downstream targets. Our therapeutic target is the transcription factor SMARCE1, which was previously identified as a key driver of invasion in early-stage breast cancer. Using a doxycycline-inducible shRNA knockdown in OVCAR8 ovarian cancer cells both in vitro and in vivo, we demonstrate that SMARCE1 is a master regulator of genes encoding proinvasive proteases in a model of human ovarian cancer. We additionally map the peptide cleavage profiles of SMARCE1-regulated proteases so as to design a readout for downstream enzymatic activity. To demonstrate the therapeutic and diagnostic potential of our approach, we engineered self-assembled layer-by-layer nanoparticles that can encapsulate nucleic acid cargo and be decorated with peptide substrates that release a urinary reporter upon exposure to SMARCE1-related proteases. In an orthotopic ovarian cancer xenograft model, theranostic nanoparticles were able to knockdown SMARCE1 which was in turn reported through a reduction in protease-activated urinary reporters. These LBL nanoparticles both silence gene products by delivering siRNA and noninvasively report on downstream target activity by delivering synthetic biomarkers to sites of disease, enabling dose-finding studies as well as longitudinal assessments of efficacy.


Subject(s)
Ovarian Neoplasms , Peptides , Humans , Female , RNA Interference , Peptides/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/therapy , Peptide Hydrolases , RNA, Small Interfering/genetics , Endopeptidases , Chromosomal Proteins, Non-Histone , DNA-Binding Proteins
3.
Nature ; 585(7826): 603-608, 2020 09.
Article in English | MEDLINE | ID: mdl-32939090

ABSTRACT

Ferroptosis-an iron-dependent, non-apoptotic cell death process-is involved in various degenerative diseases and represents a targetable susceptibility in certain cancers1. The ferroptosis-susceptible cell state can either pre-exist in cells that arise from certain lineages or be acquired during cell-state transitions2-5. However, precisely how susceptibility to ferroptosis is dynamically regulated remains poorly understood. Here we use genome-wide CRISPR-Cas9 suppressor screens to identify the oxidative organelles peroxisomes as critical contributors to ferroptosis sensitivity in human renal and ovarian carcinoma cells. Using lipidomic profiling we show that peroxisomes contribute to ferroptosis by synthesizing polyunsaturated ether phospholipids (PUFA-ePLs), which act as substrates for lipid peroxidation that, in turn, results in the induction of ferroptosis. Carcinoma cells that are initially sensitive to ferroptosis can switch to a ferroptosis-resistant state in vivo in mice, which is associated with extensive downregulation of PUFA-ePLs. We further find that the pro-ferroptotic role of PUFA-ePLs can be extended beyond neoplastic cells to other cell types, including neurons and cardiomyocytes. Together, our work reveals roles for the peroxisome-ether-phospholipid axis in driving susceptibility to and evasion from ferroptosis, highlights PUFA-ePL as a distinct functional lipid class that is dynamically regulated during cell-state transitions, and suggests multiple regulatory nodes for therapeutic interventions in diseases that involve ferroptosis.


Subject(s)
Ethers/metabolism , Ferroptosis , Peroxisomes/metabolism , Phospholipids/chemistry , Phospholipids/metabolism , Animals , CRISPR-Cas Systems/genetics , Cell Differentiation , Cell Line , Ethers/chemistry , Female , Gene Editing , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Lipid Peroxidation , Male , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Neurons/cytology , Neurons/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Peroxisomes/genetics
4.
Proc Natl Acad Sci U S A ; 119(23): e2118697119, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35648828

ABSTRACT

The blood­brain barrier represents a significant challenge for the treatment of high-grade gliomas, and our understanding of drug transport across this critical biointerface remains limited. To advance preclinical therapeutic development for gliomas, there is an urgent need for predictive in vitro models with realistic blood­brain-barrier vasculature. Here, we report a vascularized human glioblastoma multiforme (GBM) model in a microfluidic device that accurately recapitulates brain tumor vasculature with self-assembled endothelial cells, astrocytes, and pericytes to investigate the transport of targeted nanotherapeutics across the blood­brain barrier and into GBM cells. Using modular layer-by-layer assembly, we functionalized the surface of nanoparticles with GBM-targeting motifs to improve trafficking to tumors. We directly compared nanoparticle transport in our in vitro platform with transport across mouse brain capillaries using intravital imaging, validating the ability of the platform to model in vivo blood­brain-barrier transport. We investigated the therapeutic potential of functionalized nanoparticles by encapsulating cisplatin and showed improved efficacy of these GBM-targeted nanoparticles both in vitro and in an in vivo orthotopic xenograft model. Our vascularized GBM model represents a significant biomaterials advance, enabling in-depth investigation of brain tumor vasculature and accelerating the development of targeted nanotherapeutics.


Subject(s)
Blood-Brain Barrier , Brain Neoplasms , Capillary Permeability , Glioblastoma , Nanoparticles , Animals , Blood-Brain Barrier/metabolism , Brain Neoplasms/blood supply , Brain Neoplasms/metabolism , Endothelial Cells/metabolism , Glioblastoma/blood supply , Glioblastoma/metabolism , Humans , Mice , Microfluidics , Nanoparticles/metabolism , Xenograft Model Antitumor Assays
5.
Biomacromolecules ; 25(5): 2934-2952, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38687965

ABSTRACT

Localized short interfering RNA (siRNA) therapy has the potential to drive high-specificity molecular-level treatment of a variety of disease states. Unfortunately, effective siRNA therapy suffers from several barriers to its intracellular delivery. Thus, drug delivery systems that package and control the release of therapeutic siRNAs are necessary to overcome these obstacles to clinical translation. Layer-by-layer (LbL) electrostatic assembly of thin film coatings containing siRNA and protonatable, hydrolyzable poly(ß-aminoester) (PBAE) polymers is one such drug delivery strategy. However, the impact of PBAE physicochemical properties on the transfection efficacy of siRNA released from LbL thin film coatings has not been systematically characterized. In this study, we investigate the siRNA transfection efficacy of four structurally similar PBAEs in vitro. We demonstrate that small changes in structure yield large changes in physicochemical properties, such as hydrophobicity, pKa, and amine chemical structure, driving differences in the interactions between PBAEs and siRNA in polyplexes and in LbL thin film coatings for wound dressings. In our polymer set, Poly3 forms the most stable interactions with siRNA (Keff,w/w = 0.298) to slow release kinetics and enhance transfection of reporter cells in both colloidal and thin film coating approaches. This is due to its unique physiochemical properties: high hydrophobicity (clog P = 7.86), effective pKa closest to endosomal pH (pKa = 6.21), and high cooperativity in buffering (nhill = 7.2). These properties bestow Poly3 with enhanced endosomal buffering and escape properties. Taken together, this work elucidates the connections between small changes in polymer structure, emergent properties, and polyelectrolyte theory to better understand PBAE transfection efficacy.


Subject(s)
Polymers , RNA, Small Interfering , Static Electricity , RNA, Small Interfering/chemistry , RNA, Small Interfering/administration & dosage , Humans , Polymers/chemistry , Transfection/methods , Hydrophobic and Hydrophilic Interactions , Drug Delivery Systems/methods
6.
Proc Natl Acad Sci U S A ; 118(42)2021 10 19.
Article in English | MEDLINE | ID: mdl-34649991

ABSTRACT

Nanoparticle (NP) stiffness has been shown to significantly impact circulation time and biodistribution in anticancer drug delivery. In particular, the relationship between particle stiffness and tumor accumulation and penetration in vivo is an important phenomenon to consider in optimizing NP-mediated tumor delivery. Layer-by-layer (LbL) NPs represent a promising class of multifunctional nanoscale drug delivery carriers. However, there has been no demonstration of the versatility of LbL systems in coating systems with different stiffnesses, and little is known about the potential role of LbL NP stiffness in modulating in vivo particle trafficking, although NP modulus has been recently studied for its impact on pharmacokinetics. LbL nanotechnology enables NPs to be functionalized with uniform coatings possessing molecular tumor-targeting properties, independent of the NP core stiffness. Here, we report that the stiffness of LbL NPs is directly influenced by the mechanical properties of its underlying liposomal core, enabling the modulation and optimization of LbL NP stiffness while preserving LbL NP outer layer tumor-targeting and stealth properties. We demonstrate that the stiffness of LbL NPs has a direct impact on NP pharmacokinetics, organ and tumor accumulation, and tumor penetration-with compliant LbL NPs having longer elimination half-life, higher tumor accumulation, and higher tumor penetration. Our findings underscore the importance of NP stiffness as a design parameter in enhancing the delivery of LbL NP formulations.


Subject(s)
Nanoparticles/chemistry , Neoplasms/metabolism , Cell Line, Tumor , Drug Delivery Systems , Half-Life , Humans , Liposomes , Polymers/chemistry , Tissue Distribution
7.
Soft Matter ; 19(16): 3033-3046, 2023 Apr 26.
Article in English | MEDLINE | ID: mdl-37038739

ABSTRACT

Cationic poly(amido amine) (PAMAM) dendrimers exhibit great potential for use in drug delivery, but their high charge density leads to an inherent cytotoxicity. To increase biocompatibility, many studies have attached poly(ethylene glycol) (PEG) chains to the dendrimer surface. It is unclear how these tethered PEG chains influence the physicochemical properties of the dendrimer. Here, we develop a fluorescence-based assay utilizing anionic biological tissue to quantify the electrostatic binding affinity of a library of PEG-PAMAM conjugates with various PEG chain lengths and grafting densities. We find that covalently bound PEG chains reduce the electrostatic binding affinity more significantly than what can be achieved through covalent bonds only. Contrary to previous thought, this reduction is not explained by the steric hindrance effects of PEG chains, suggesting that other, non-covalent interactions between PEG and PAMAM are present. Using acetylated PAMAM conjugates, we convert electrostatic binding affinity to the number of charged amines accessible to the physiological environment. These data, coupled with 1H-NMR, allows us to study more closely the non-covalent interactions between PEG and PAMAM. We find that increasing PEG chain length increases the number of non-covalent interactions. Additionally, at low grafting densities, increasing the number of PEG chains on the PAMAM surface also increases the non-covalent interactions. At higher grafting densities, however, PEG chains sterically repel one another, forcing chains to elongate away from the surface and reducing the number of interactions between PAMAM and individual PEG chains. The data presented here provides a framework for a more precise mechanistic understanding of how the length and density of tethered PEG chains on PAMAM dendrimers influence drug delivery properties.


Subject(s)
Dendrimers , Dendrimers/chemistry , Drug Delivery Systems , Polyethylene Glycols/chemistry
8.
Proc Natl Acad Sci U S A ; 117(49): 31376-31385, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33229526

ABSTRACT

For a myriad of different reasons most antimicrobial peptides (AMPs) have failed to reach clinical application. Different AMPs have different shortcomings including but not limited to toxicity issues, potency, limited spectrum of activity, or reduced activity in situ. We synthesized several cationic peptide mimics, main-chain cationic polyimidazoliums (PIMs), and discovered that, although select PIMs show little acute mammalian cell toxicity, they are potent broad-spectrum antibiotics with activity against even pan-antibiotic-resistant gram-positive and gram-negative bacteria, and mycobacteria. We selected PIM1, a particularly potent PIM, for mechanistic studies. Our experiments indicate PIM1 binds bacterial cell membranes by hydrophobic and electrostatic interactions, enters cells, and ultimately kills bacteria. Unlike cationic AMPs, such as colistin (CST), PIM1 does not permeabilize cell membranes. We show that a membrane electric potential is required for PIM1 activity. In laboratory evolution experiments with the gram-positive Staphylococcus aureus we obtained PIM1-resistant isolates most of which had menaquinone mutations, and we found that a site-directed menaquinone mutation also conferred PIM1 resistance. In similar experiments with the gram-negative pathogen Pseudomonas aeruginosa, PIM1-resistant mutants did not emerge. Although PIM1 was efficacious as a topical agent, intraperitoneal administration of PIM1 in mice showed some toxicity. We synthesized a PIM1 derivative, PIM1D, which is less hydrophobic than PIM1. PIM1D did not show evidence of toxicity but retained antibacterial activity and showed efficacy in murine sepsis infections. Our evidence indicates the PIMs have potential as candidates for development of new drugs for treatment of pan-resistant bacterial infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Designer Drugs/pharmacology , Imidazoles/pharmacology , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Cell Death/drug effects , Cell Line , Cell Membrane/drug effects , Designer Drugs/chemistry , Designer Drugs/therapeutic use , Humans , Hydrophobic and Hydrophilic Interactions , Imidazoles/chemistry , Imidazoles/therapeutic use , Membrane Potentials/drug effects , Mice , Microbial Sensitivity Tests , Microbial Viability/drug effects , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , Pseudomonas Infections/pathology , Pseudomonas aeruginosa/drug effects , Sepsis/drug therapy , Sepsis/prevention & control , Skin/drug effects , Skin/microbiology , Skin/pathology
9.
Chem Eng J ; 4642023 May 15.
Article in English | MEDLINE | ID: mdl-38737525

ABSTRACT

Immune stimulating complexes (ISCOMs) are safe and effective saponin-based adjuvants formed by the self-assembly of saponin, cholesterol, and phospholipids in water to form cage-like 30-40 nm diameter particles. Inclusion of the Toll-like receptor 4 agonist monophosphoryl lipid A (MPLA) in ISCOM particles yields a promising next-generation adjuvant termed Saponin-MPLA NanoParticles (SMNP). In this work, we detail protocols to produce ISCOMs or SMNP via a tangential flow filtration (TFF) process suitable for scalable synthesis and Good Manufacturing Practice (GMP) production of clinical-grade adjuvants. SMNP or ISCOM components were solubilized in micelles of the surfactant MEGA-10, then diluted below the critical micelle concentration (CMC) of the surfactant to drive ISCOM self-assembly. Assembly of ISCOM/SMNP particles using the purified saponin QS-21 used in clinical-grade saponin adjuvants was found to require controlled stepwise dilution of the initial micellar solution, to prevent formation of undesirable kinetically-trapped aggregate species. An optimized protocol gave yields of ~77% based on the initial feed of QS-21 and the final SMNP particle composition mirrored the feed ratios of the components. Further, samples were highly homogeneous with comparable quality to that of material prepared at lab scale by dialysis and purified via size-exclusion chromatography. This protocol may be useful for clinical preparation of ISCOM-based vaccine adjuvants and therapeutics.

10.
Bioconjug Chem ; 33(11): 2065-2075, 2022 11 16.
Article in English | MEDLINE | ID: mdl-36282941

ABSTRACT

Nanoparticle (NP) drug carriers have revolutionized medicine and increased patient quality of life. Clinically approved formulations typically succeed because of reduced off-target toxicity of the cargo. However, increasing carrier accumulation at disease sites through precise targeting remains one of the biggest challenges in the field. Novel multivalent ligand presentations and self-assembled constructs can enhance cell association, but an inability to draw direct comparisons across formulations has hindered progress. Furthermore, how nanoparticle structure influences function often is unclear. In this report, we leverage the well-characterized hyaluronic acid (HA)-CD44 binding pair to investigate how the surface architecture of modified NPs impacts their association with ovarian cancer cells that overexpress CD44. We functionalized anionic liposomes with 5 kDa HA by either covalent conjugation via surface coupling or electrostatic self-assembly using the layer-by-layer (LbL) adsorption method. Comparing these two methods, we observed a consistent enhancement of NP-cell association with the self-assembly LbL technique, particularly with higher molecular weight (≥10 kDa) HA. To further optimize association, we increased the surface-available HA. We synthesized a bottlebrush glycopolymer composed of a polynorbornene backbone and pendant 5 kDa HA and layered this macromolecule onto NPs. Flow cytometry revealed that the LbL HA bottlebrush NP outperformed the LbL linear display of HA. Cellular visualization by deconvolution optical microscopy corroborated results from all three constructs. Using exogenous HA to block NP-CD44 interactions, we found the LbL HA bottlebrush NP had a 4-fold higher binding avidity than the best-performing LbL linear HA NP. We further observed that decreasing the density of HA bottlebrush side chains to 75% had minimal impact on LbL NP stability or cell association, though we did see a reduction in binding avidity with this side-chain-modified NP. Our studies indicate that LbL surfaces are highly effective for multivalent displays, and the mode in which they present a targeting ligand can be optimized for NP cell targeting.


Subject(s)
Hyaluronic Acid , Nanoparticles , Humans , Hyaluronic Acid/chemistry , Ligands , Quality of Life , Nanoparticles/chemistry , Hyaluronan Receptors/metabolism , Drug Carriers/chemistry , Cell Line, Tumor
11.
Proc Natl Acad Sci U S A ; 116(33): 16473-16478, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31358641

ABSTRACT

Sustained exposure of lymphoid tissues to vaccine antigens promotes humoral immunity, but traditional bolus immunizations lead to rapid antigen clearance. We describe a technology to tailor vaccine kinetics in a needle-free platform translatable to human immunization. Solid pyramidal microneedle (MN) arrays were fabricated with silk fibroin protein tips encapsulating a stabilized HIV envelope trimer immunogen and adjuvant, supported on a dissolving polymer base. Upon brief skin application, vaccine-loaded silk tips are implanted in the epidermis/upper dermis where they release vaccine over a time period determined by the crystallinity of the silk matrix. Following MN immunization in mice, Env trimer was released over 2 wk in the skin, correlating with increased germinal center (GC) B cell responses, a ∼1,300-fold increase in serum IgG titers and a 16-fold increase in bone marrow (BM) plasma cells compared with bolus immunization. Thus, implantable MNs provide a practical means to substantially enhance humoral immunity to subunit vaccines.


Subject(s)
Delayed-Action Preparations/pharmacology , Immunity, Humoral , Needles , Prostheses and Implants , Vaccination , Animals , Antibody Formation/immunology , Antigens/immunology , Bombyx , Germinal Center/immunology , Lymph Nodes/immunology , Mice, Inbred BALB C , Silk , Skin
12.
Annu Rev Biomed Eng ; 22: 1-24, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32084319

ABSTRACT

Controlled drug delivery formulations have revolutionized treatments for a range of health conditions. Over decades of innovation, layer-by-layer (LbL) self-assembly has emerged as one of the most versatile fabrication methods used to develop multifunctional controlled drug release coatings. The numerous advantages of LbL include its ability to incorporate and preserve biological activity of therapeutic agents; coat multiple substrates of all scales (e.g., nanoparticles to implants); and exhibit tuned, targeted, and/or responsive drug release behavior. The functional behavior of LbL films can be related to their physicochemical properties. In this review, we highlight recent advances in the development of LbL-engineered biomaterials for drug delivery, demonstrating their potential in the fields of cancer therapy, microbial infection prevention and treatment, and directing cellular responses. We discuss the various advantages of LbL biomaterial design for a given application as demonstrated through in vitro and in vivo studies.


Subject(s)
Biocompatible Materials , Drug Delivery Systems , Nanoparticles/chemistry , Neoplasms/drug therapy , Animals , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Coated Materials, Biocompatible , Electrolytes , Hydrogen-Ion Concentration , In Vitro Techniques , Mice , Pharmaceutical Preparations , Precision Medicine , Regeneration , Surface Properties
13.
Proc Natl Acad Sci U S A ; 115(12): E2696-E2705, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29432194

ABSTRACT

Small interfering RNA (siRNA) represents a promising class of inhibitors in both fundamental research and the clinic. Numerous delivery vehicles have been developed to facilitate siRNA delivery. Nevertheless, achieving highly potent RNA interference (RNAi) toward clinical translation requires efficient formation of RNA-induced gene-silencing complex (RISC) in the cytoplasm. Here we coencapsulate siRNA and the central RNAi effector protein Argonaute 2 (Ago2) via different delivery carriers as a platform to augment RNAi. The physical clustering between siRNA and Ago2 is found to be indispensable for enhanced RNAi. Moreover, by utilizing polyamines bearing the same backbone but distinct cationic side-group arrangements of ethylene diamine repeats as the delivery vehicles, we find that the molecular structure of these polyamines modulates the degree of siRNA/Ago2-mediated improvement of RNAi. We apply this strategy to silence the oncogene STAT3 and significantly prolong survival in mice challenged with melanoma. Our findings suggest a paradigm for RNAi via the synergistic coassembly of RNA with helper proteins.


Subject(s)
Argonaute Proteins/genetics , Genetic Therapy/methods , RNA Interference , RNA, Small Interfering/administration & dosage , RNA-Induced Silencing Complex/chemistry , Animals , Argonaute Proteins/metabolism , Drug Delivery Systems/methods , Melanoma, Experimental/genetics , Melanoma, Experimental/mortality , Melanoma, Experimental/therapy , Mice, Inbred C57BL , Oncogenes/genetics , Polyamines/chemistry , RNA, Antisense/administration & dosage , RNA, Antisense/pharmacology , RNA, Double-Stranded/administration & dosage , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/genetics , RNA, Messenger , RNA, Small Interfering/chemistry , RNA-Induced Silencing Complex/genetics , RNA-Induced Silencing Complex/metabolism , STAT3 Transcription Factor/genetics , Structure-Activity Relationship , Transfection/methods
14.
Bioconjug Chem ; 31(9): 2211-2219, 2020 09 16.
Article in English | MEDLINE | ID: mdl-32786506

ABSTRACT

We report the surface functionalization of anionic layer by layer nanoparticles (LbL NPs) with cationic tumor-penetrating peptides (TPPs) via electrostatic adsorption while retaining particle stability and charge characteristics. This strategy eliminates the need for structural modifications of the peptide and enables facile functionalization of surface chemistries difficult to modify or inaccessible via covalent conjugation strategies. We show that both carboxylated and sulfated LbL NPs are able to accommodate linear and cyclic TPPs and used fluorescence-based detection assays to quantify peptide loading per NP. We also demonstrate that TPP activity is retained upon adsorption, implying sufficient numbers of peptides take on the appropriate surface orientation, enabling efficient uptake of functionalized NPs in vitro, as characterized via flow cytometry and deconvolution microscopy. Overall, we believe that this strategy will serve as a broadly applicable approach to impart electrostatically assembled NPs with bioactive peptide motifs.


Subject(s)
Cell-Penetrating Peptides/chemistry , Nanoparticles/chemistry , Adsorption , Cell Line, Tumor , Cell-Penetrating Peptides/metabolism , Humans , Liposomes/chemistry , Liposomes/metabolism , Nanoparticles/metabolism , Static Electricity , Surface Properties
15.
Biomacromolecules ; 21(2): 566-580, 2020 02 10.
Article in English | MEDLINE | ID: mdl-31846304

ABSTRACT

Expanding the toolkit of modular and functional synthetic material systems for biomimetic extracellular matrices (ECMs) is needed for achieving more predictable and characterizable cell culture. In the present study, we engineered a synthetic hydrogel system incorporating poly(γ-propargyl-l-glutamate) (PPLG), an N-carboxy anhydride polypeptide with a unique α-helical secondary structure. PPLG macromers were cross-linked into poly(ethylene glycol) (PEG) networks to form hybrid polypeptide-PEG hydrogels. We compared the properties of PPLG-PEG to systems where the PPLG macromers were replaced with 8-arm PEG or poly(γ-propargyl-d,l-glutamate) (PPDLG), which has a flexible random-coil conformation. We evaluated each hydrogel system as synthetic ECMs for two-dimensional (2D) endothelial cell culture. Cells on PPLG-PEG displayed superior attachment and spreading at comparable adhesion ligand incorporation concentrations, demonstrating the unique benefit of combining the more rigid and hydrophobic α-helical PPLG within the more flexible and hydrophilic PEG matrix. The modular PPLG macromer is a promising building block for developing other types of PPLG-based hydrogels with favorable and tunable properties.


Subject(s)
Cell Culture Techniques/instrumentation , Extracellular Matrix/chemistry , Hydrogels/chemistry , Peptides/chemistry , Cell Adhesion , Cell Culture Techniques/methods , Cells, Cultured , Fluorescence Recovery After Photobleaching , Humans , Hydrophobic and Hydrophilic Interactions , Induced Pluripotent Stem Cells/cytology , Permeability
16.
Angew Chem Int Ed Engl ; 59(7): 2776-2783, 2020 02 10.
Article in English | MEDLINE | ID: mdl-31747099

ABSTRACT

Layer-by-layer nanoparticles (NPs) are modular drug delivery vehicles that incorporate multiple functional materials through sequential deposition of polyelectrolytes onto charged nanoparticle cores. Herein, we combined the multicomponent features and tumor targeting capabilities of layer-by-layer assembly with functional biosensing peptides to create a new class of nanotheranostics. These NPs encapsulate a high weight percentage of siRNA while also carrying a synthetic biosensing peptide on the surface that is cleaved into a urinary reporter upon exposure to specific proteases overexpressed in the tumor microenvironment. Importantly, this biosensor reports back on a molecular signature characteristic to metastatic tumors and associated with poor prognosis, MMP9 protease overexpression. This nanotheranostic mediates noninvasive urinary-based diagnostics in mouse models of three different cancers with simultaneous gene silencing in flank and metastatic mouse models of ovarian cancer.


Subject(s)
Colorectal Neoplasms/diagnosis , Nanoparticles/chemistry , Ovarian Neoplasms/diagnosis , Peptides/chemistry , Theranostic Nanomedicine , Animals , Biosensing Techniques , Colorectal Neoplasms/genetics , Drug Delivery Systems , Female , Gene Silencing , Mice , Ovarian Neoplasms/genetics , Peptides/chemical synthesis
17.
Adv Funct Mater ; 29(20)2019 May 16.
Article in English | MEDLINE | ID: mdl-31839764

ABSTRACT

Using siRNA therapeutics to treat hematologic malignancies has been unsuccessful because blood cancer cells exhibit remarkable resistance to standard transfection methods. Herein we report the successful delivery of siRNA therapeutics with a dual-targeted, layer-by-layer nanoparticle (LbL-NP). The LbL-NP protects siRNA from nucleases in the bloodstream by embedding it within polyelectrolyte layers that coat a polymeric core. The outermost layer consists of hyaluronic acid (a CD44-ligand) covalently conjugated to CD20 antibodies. The CD20/CD44 dual-targeting outer layer provides precise binding to blood cancer cells, followed by receptor-mediated endocytosis of the LbL-NP. We use this siRNA delivery platform to silence B-cell lymphoma 2 (BCL-2), a pro-survival protein, in vitro and in vivo. The dual-targeting approach significantly enhanced internalization of BCL-2 siRNA in lymphoma and leukemia cells, which led to significant downregulation of BCL-2 expression. Systemic administration of the dual-targeted, siRNA-loaded nanoparticle induced apoptosis and hampered proliferation of blood cancer cells both in cell culture and in orthotopic non-Hodgkin's lymphoma animal models. These results provide the basis for approaches to targeting blood-borne cancers and other diseases, and suggest that LbL nanoassemblies are a promising approach for delivering therapeutic siRNA to hematopoetic cell types that are known to evade transfection by other means.

18.
Proc Natl Acad Sci U S A ; 113(19): 5179-84, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27114520

ABSTRACT

Fluorescence imaging in the second near-infrared window (NIR-II, 1,000-1,700 nm) features deep tissue penetration, reduced tissue scattering, and diminishing tissue autofluorescence. Here, NIR-II fluorescent probes, including down-conversion nanoparticles, quantum dots, single-walled carbon nanotubes, and organic dyes, are constructed into biocompatible nanoparticles using the layer-by-layer (LbL) platform due to its modular and versatile nature. The LbL platform has previously been demonstrated to enable incorporation of diagnostic agents, drugs, and nucleic acids such as siRNA while providing enhanced blood plasma half-life and tumor targeting. This work carries out head-to-head comparisons of currently available NIR-II probes with identical LbL coatings with regard to their biodistribution, pharmacokinetics, and toxicities. Overall, rare-earth-based down-conversion nanoparticles demonstrate optimal biological and optical performance and are evaluated as a diagnostic probe for high-grade serous ovarian cancer, typically diagnosed at late stage. Successful detection of orthotopic ovarian tumors is achieved by in vivo NIR-II imaging and confirmed by ex vivo microscopic imaging. Collectively, these results indicate that LbL-based NIR-II probes can serve as a promising theranostic platform to effectively and noninvasively monitor the progression and treatment of serous ovarian cancer.


Subject(s)
Fluorescent Dyes/chemical synthesis , Microscopy, Fluorescence/methods , Nanocapsules/chemistry , Ovarian Neoplasms/diagnostic imaging , Spectroscopy, Near-Infrared/methods , Animals , Cell Line, Tumor , Contrast Media/chemical synthesis , Contrast Media/pharmacokinetics , Crystallization/methods , Female , Fluorescent Dyes/pharmacokinetics , Humans , Mice , Mice, Inbred BALB C , Nanocapsules/ultrastructure , Organ Specificity , Ovarian Neoplasms/pathology , Reproducibility of Results , Sensitivity and Specificity , Theranostic Nanomedicine/methods , Tissue Distribution
19.
Nucleic Acids Res ; 44(2): 545-57, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26704983

ABSTRACT

Large dsRNA molecules can cause potent cytotoxic and immunostimulatory effects through the activation of pattern recognition receptors; however, synthetic versions of these molecules are mostly limited to simple sequences like poly-I:C and poly-A:U. Here we show that large RNA molecules generated by rolling circle transcription fold into periodic-shRNA (p-shRNA) structures and cause potent cytotoxicity and gene silencing when delivered to cancer cells. We determined structural requirements for the dumbbell templates used to synthesize p-shRNA, and showed that these molecules likely adopt a co-transcriptionally folded structure. The cytotoxicity of p-shRNA was robustly observed across four different cancer cell lines using two different delivery systems. Despite having a considerably different folded structure than conventional dsRNA, the cytotoxicity of p-shRNA was either equal to or substantially greater than that of poly-I:C depending on the delivery vehicle. Furthermore, p-shRNA caused greater NF-κB activation in SKOV3 cells compared to poly-I:C, indicating that it is a powerful activator of innate immunity. The tuneable sequence and combined gene silencing, immunostimulatory and cytotoxic capacity of p-shRNA make it an attractive platform for cancer immunotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , RNA Interference , RNA, Small Interfering/pharmacology , Antineoplastic Agents/immunology , Antineoplastic Agents/metabolism , Base Sequence , Caspase 3/genetics , Caspase 3/immunology , Caspase 7/genetics , Caspase 7/immunology , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Humans , Immunity, Innate , Luciferases/antagonists & inhibitors , Luciferases/genetics , Luciferases/immunology , Molecular Sequence Data , NF-kappa B/biosynthesis , NF-kappa B/metabolism , Nucleic Acid Conformation , Poly I-C/genetics , Poly I-C/immunology , Poly I-C/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Transcription, Genetic
20.
J Am Chem Soc ; 139(7): 2741-2749, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28133963

ABSTRACT

The detection of rare circulating tumor cells (CTCs) in the blood of cancer patients has the potential to be a powerful and noninvasive method for examining metastasis, evaluating prognosis, assessing tumor sensitivity to drugs, and monitoring therapeutic outcomes. In this study, we have developed an efficient strategy to isolate CTCs from the blood of breast cancer patients using a microfluidic immune-affinity approach. Additionally, to gain further access to these rare cells for downstream characterization, our strategy allows for easy detachment of the captured CTCs from the substrate without compromising cell viability or the ability to employ next generation RNA sequencing for the identification of specific breast cancer genes. To achieve this, a chemical ligand-exchange reaction was engineered to release cells attached to a gold nanoparticle coating bound to the surface of a herringbone microfluidic chip (NP-HBCTC-Chip). Compared to the use of the unmodified HBCTC-Chip, our approach provides several advantages, including enhanced capture efficiency and recovery of isolated CTCs.


Subject(s)
Gold/chemistry , Metal Nanoparticles/chemistry , Microfluidic Analytical Techniques , Neoplastic Cells, Circulating/chemistry , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Adhesion , Cell Line, Tumor , Female , Fluorescent Antibody Technique , Humans , Ligands , Surface Properties , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL