Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
ACS Nano ; 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38870478

ABSTRACT

Amidst the rapid growth of protein therapeutics as a drug class, there is an increased focus on designing systems to effectively deliver proteins to target organs. Quantitative monitoring of protein distributions in tissues is essential for optimal development of delivery systems; however, existing strategies can have limited accuracy, making it difficult to assess suborgan dosing. Here, we describe a quantitative imaging approach that utilizes metal-coded mass tags and laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS) to quantify the suborgan distributions of proteins in tissues that have been delivered by polymeric nanocarriers. Using this approach, we measure nanomole per gram levels of proteins as delivered by guanidinium-functionalized poly(oxanorborneneimide) (PONI) polymers to various tissues, including the alveolar region of the lung. Due to the multiplexing capability of the LA-ICP-MS imaging, we are also able to simultaneously quantify protein and polymer distributions, obtaining valuable information about the relative excretion pathways of the protein cargo and carrier. This imaging approach will facilitate quantitative correlations between nanocarrier properties and protein cargo biodistributions.

2.
Mater Horiz ; 10(12): 5500-5507, 2023 11 27.
Article in English | MEDLINE | ID: mdl-37815454

ABSTRACT

Treatment of wound biofilm infections faces challenges from both pathogens and uncontrolled host immune response. Treating both issues through a single vector would provide enhanced wound healing. Here, we report the use of a potent cationic antimicrobial polymer to generate siRNA polyplexes for dual-mode treatment of wound biofilms in vivo. These polyplexes act both as an antibiofilm agent and a delivery vehicle for siRNA for the knockdown of biofilm-associated pro-inflammatory MMP9 in host macrophages. The resulting polyplexes were effective in vitro, eradicating MRSA biofilms and efficiently delivering siRNA to macrophages in vitro with concomitant knockdown of MMP9. These polyplexes were likewise effective in an in vivo murine wound biofilm model, significantly reducing bacterial load in the wound (∼99% bacterial clearance) and reducing MMP9 expression by 80% (qRT-PCR). This combination therapeutic strategy dramatically reduced wound purulence and significantly expedited wound healing. Taken together, these polyplexes provide an effective and translatable strategy for managing biofilm-infected wounds.


Subject(s)
Anti-Infective Agents , Matrix Metalloproteinase 9 , Animals , Mice , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Wound Healing/genetics , Biofilms
3.
J Control Release ; 362: 513-523, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37666301

ABSTRACT

Integration of antimicrobial polymeric nanoparticles into hydrogel materials presents a promising strategy to address multidrug-resistant biofilm infections. Here we report an injectable hydrogel loaded with engineered cationic antimicrobial polymeric nanoparticles (PNPs) for the effective topical treatment of severe wound biofilm infections. The PNPs demonstrated biofilm penetration and disruption, resulting in the eradication of resistant and persister cells that reside within the biofilm. Significantly, PNPs did not elicit resistance development even after multiple exposures to sub-therapeutic doses. In vitro studies showed PNPs significantly reduced prolonged inflammation due to infection and promoted fibroblast migration. These PNPs were then incorporated into Poloxamer 407 (P407) hydrogels and utilized as an inert carrier for PNPs to provide a controlled and sustained topical release of the antimicrobial nanoparticles at the wound area. In vivo studies using a mature (4-day) wound biofilm infection in a murine model mimicking severe human wound infections demonstrated provided 99% bacterial biofilm clearance and significantly enhanced wound healing. Overall, this work demonstrated the efficacy and selectivity of the antimicrobial polymer-loaded hydrogel platform as a topical treatment for difficult-to-treat wound biofilm infections.

4.
Anal Chem ; 95(32): 12177-12183, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37535805

ABSTRACT

Macrophages are key components of the innate immune system that have essential functions in physiological processes and diseases. The phenotypic plasticity of macrophages allows cells to be polarized into a multidimensional spectrum of phenotypes, broadly classed as pro-inflammatory (M1) and anti-inflammatory (M2) states. Repolarization of M1 to M2 phenotypes alters the immune response to ameliorate autoimmune and inflammation-associated diseases. Detection of this repolarization, however, is challenging to execute in high-throughput applications. In this work, we demonstrate the ability of a single polymer fabricated to provide a six-channel sensor array that can determine macrophage polarization phenotypes. This sensing platform provides a sensitive and high-throughput tool for detecting drug-induced M1-to-M2 repolarization, allowing the identification of new therapeutic leads for inflammatory diseases. The ability of this sensor array to discriminate different M2 subtypes induced by drugs can also improve the efficacy evaluation of anti-inflammatory drugs and avoid adverse effects.


Subject(s)
Anti-Inflammatory Agents , Macrophages , Humans , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Inflammation/drug therapy , Phenotype
5.
Nanoscale ; 15(24): 10351-10359, 2023 Jun 23.
Article in English | MEDLINE | ID: mdl-37288531

ABSTRACT

Vaccination through cellular transfection of nucleotide-based vaccines is a powerful approach to combatting disease. Plasmid DNA (pDNA) vaccines are particularly promising vectors for non-viral immunomodulation that afford high degrees of potency and flexibility. Versatile guanidinium-functionalized poly(oxanorbornene)imide (PONI-Guan) homopolymers were used to facilitate non-disruptive pDNA condensation into discrete polyplexes, enabling efficient in vitro transfection of endothelial cells and HD-11 macrophages. Translation of these vectors for vaccination of white leghorn chickens against Newcastle disease virus (NDV) elicited strong humoral immune responses against the virus. This approach presents a highly versatile method for targeted immunomodulation in vivo, with the potential for translatability as a non-viral vaccine platform.


Subject(s)
Chickens , Polymers , Animals , Chickens/genetics , Endothelial Cells , Plasmids/genetics , DNA/genetics , Vaccination
6.
J Control Release ; 357: 31-39, 2023 05.
Article in English | MEDLINE | ID: mdl-36948419

ABSTRACT

Bioorthogonal catalysis via transition metal catalysts (TMCs) enables the generation of therapeutics locally through chemical reactions not accessible by biological systems. This localization can enhance the efficacy of anticancer treatment while minimizing off-target effects. The encapsulation of TMCs into nanomaterials generates "nanozymes" to activate imaging and therapeutic agents. Here, we report the use of cationic bioorthogonal nanozymes to create localized "drug factories" for cancer therapy in vivo. These nanozymes remained present at the tumor site at least seven days after a single injection due to the interactions between cationic surface ligands and negatively charged cell membranes and tissue components. The prodrug was then administered systemically, and the nanozymes continuously converted the non-toxic molecules into active drugs locally. This strategy substantially reduced the tumor growth in an aggressive breast cancer model, with significantly reduced liver damage compared to traditional chemotherapy.


Subject(s)
Breast Neoplasms , Nanostructures , Humans , Female , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Diagnostic Imaging , Catalysis , Cell Membrane
7.
ACS Nano ; 17(5): 4315-4326, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36802503

ABSTRACT

Uncontrolled inflammation is responsible for acute and chronic diseases in the lung. Regulating expression of pro-inflammatory genes in pulmonary tissue using small interfering RNA (siRNA) is a promising approach to combatting respiratory diseases. However, siRNA therapeutics are generally hindered at the cellular level by endosomal entrapment of delivered cargo and at the organismal level by inefficient localization in pulmonary tissue. Here we report efficient anti-inflammatory activity in vitro and in vivo using polyplexes of siRNA and an engineered cationic polymer (PONI-Guan). PONI-Guan/siRNA polyplexes efficiently deliver siRNA cargo to the cytosol for highly efficient gene knockdown. Significantly, these polyplexes exhibit inherent targeting to inflamed lung tissue following intravenous administration in vivo. This strategy achieved effective (>70%) knockdown of gene expression in vitro and efficient (>80%) silencing of TNF-α expression in lipopolysaccharide (LPS)-challenged mice using a low (0.28 mg/kg) siRNA dosage.


Subject(s)
Pneumonia , Polymers , Animals , Mice , RNA, Small Interfering , Polymers/metabolism , RNA, Double-Stranded/metabolism , Endosomes/metabolism , Pneumonia/therapy , Pneumonia/metabolism
8.
Pharmaceutics ; 15(1)2023 Jan 08.
Article in English | MEDLINE | ID: mdl-36678847

ABSTRACT

Current intracellular protein delivery strategies face the challenge of endosomal entrapment and consequent degradation of protein cargo. Methods to efficiently deliver proteins directly to the cytosol have the potential to overcome this hurdle. Here, we report the use of a straightforward approach of protein modification using citraconic anhydride to impart an overall negative charge on the proteins, enabling them to assemble with positively charged nano vectors. This strategy uses anhydride-modified proteins to electrostatically form polymer-protein nanocomposites with a cationic guanidinium-functionalized polymer. These supramolecular self-assemblies demonstrated the efficient cytosolic delivery of modified proteins through a membrane fusion-like mechanism. This approach was validated on five cell lines and seven proteins as cargo. Retention of protein function was confirmed through efficient cell killing via the intracellular enzymatic activity of RNase A. This platform provides a versatile, straightforward, and single-step method of protein modification and efficient direct cytosolic protein delivery.

9.
ACS Nano ; 16(5): 7323-7330, 2022 05 24.
Article in English | MEDLINE | ID: mdl-35435664

ABSTRACT

Current strategies for the delivery of proteins into cells face general challenges of endosomal entrapment and concomitant degradation of protein cargo. Efficient delivery directly to the cytosol overcomes this obstacle: we report here the use of biotin-streptavidin tethering to provide a modular approach to the generation of nanovectors capable of a cytosolic delivery of biotinylated proteins. This strategy uses streptavidin to organize biotinylated protein and biotinylated oligo(glutamate) peptide into modular complexes that are then electrostatically self-assembled with a cationic guanidinium-functionalized polymer. The resulting polymer-protein nanocomposites demonstrate efficient cytosolic delivery of six biotinylated protein cargos of varying size, charge, and quaternary structure. Retention of protein function was established through efficient cell killing via delivery of the chemotherapeutic enzyme granzyme A. This platform represents a versatile and modular approach to intracellular delivery through the noncovalent tethering of multiple components into a single delivery vector.


Subject(s)
Biotin , Nanocomposites , Streptavidin/chemistry , Biotin/chemistry , Cytosol/metabolism , Proteins/chemistry , Polymers/chemistry
10.
Pharm Res ; 39(6): 1197-1204, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35297498

ABSTRACT

PURPOSE: Cytosolic delivery of proteins accesses intracellular targets for chemotherapy and immunomodulation. Current delivery systems utilize inefficient endosomal pathways of uptake and escape that lead to degradation of delivered cargo. Cationic poly(oxanorbornene)imide (PONI) polymers enable highly efficient cytosolic delivery of co-engineered proteins, but aggregation and denaturation in solution limits shelf life. In the present study we evaluate polymer-protein nanocomposite vehicles as candidates for lyophilization and point-of-care resuspension to provide a transferrable technology for cytosolic protein delivery. METHODS: Self-assembled nanocomposites of engineered poly(glutamate)-tagged (E-tagged) proteins and guanidinium-functionalized PONI homopolymers were generated, lyophilized, and stored for 2 weeks. After reconstitution and delivery, cytosolic access of E-tagged GFP cargo (GFPE15) was assessed through diffuse cytosolic and nuclear fluorescence, and cell killing with chemotherapeutic enzyme Granzyme A (GrAE10). Efficiency was quantified between freshly prepared and lyophilized samples. RESULTS: Reconstituted nanocomposites retained key structural features of freshly prepared assemblies, with minimal loss of material. Cytosolic delivery (> 80% efficiency of freshly prepared nanocomposites) of GFPE15 was validated in several cell lines, with intracellular access validated and quantified through diffusion into the nucleus. Delivery of GrAE10 elicited significant tumorigenic cell death. Intracellular access of cytotoxic protein was validated through cell viability. CONCLUSION: Reconstituted nanocomposites achieved efficient cytosolic delivery of protein cargo and demonstrated therapeutic applicability with delivery of GrAE10. Overall, this strategy represents a versatile and highly translatable method for cytosolic delivery of proteins.


Subject(s)
Polymers , Proteins , Cytosol/metabolism , Endosomes/metabolism , Freeze Drying , Polymers/chemistry , Proteins/chemistry
11.
Mater Horiz ; 8(12): 3424-3431, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34700339

ABSTRACT

Bioorthogonal transformation of imaging and therapeutic substrates using transition metal catalysts (TMCs) provides a toolkit with diverse applications in biomedicine. Controlled localization of bioorthogonal catalysis is key for enhancing their therapeutic efficacy by minimizing off-target effects. Red blood cells (RBCs) are highly biocompatible and are susceptible to hemolysis by bacterial toxins, providing them with intrinsic targeting to bacterial infections. A hitchhiking strategy using RBCs is reported, that activates bioorthogonal catalysis at infection sites. A library of nanoparticles embedded with TMCs (nanozymes) featuring diverse functional groups with different binding ability to RBCs is generated. These engineered nanozymes bind to RBCs and subsequently release upon hemolysis by bacterial toxins, resulting in selective accumulation at the site of bacterial infections. The antimicrobial action is specific: catalytic activation of pro-antibiotics eradicated pathogenic biofilms without harming non-virulent bacterial species.


Subject(s)
Bacterial Infections , Nanoparticles , Transition Elements , Bacterial Infections/drug therapy , Catalysis , Erythrocytes , Humans
12.
Bioconjug Chem ; 32(5): 891-896, 2021 05 19.
Article in English | MEDLINE | ID: mdl-33872490

ABSTRACT

Intracellular protein delivery is a transformative tool for biologics research and medicine. Delivery into the cytosol allows proteins to diffuse throughout the cell and access subcellular organelles. Inefficient delivery caused by endosomal entrapment is often misidentified as cytosolic delivery. This inaccuracy muddles what should be a key checkpoint in assessing delivery efficiency. Green fluorescent protein (GFP) is a robust cargo small enough to passively diffuse from the cytosol into the nucleus. Fluorescence of GFP in the nucleus is a direct readout for cytosolic access and effective delivery. Here, we highlight recent examples from the literature for the accurate assessment of cytosolic protein delivery using GFP fluorescence in the cytosol and nucleus.


Subject(s)
Bacterial Proteins/metabolism , Cell Nucleus/metabolism , Cytosol/metabolism , Luminescent Proteins/metabolism , Active Transport, Cell Nucleus , Animals , Humans
13.
Cancers (Basel) ; 13(5)2021 Mar 02.
Article in English | MEDLINE | ID: mdl-33801424

ABSTRACT

Big data analysis has revealed the upregulation of cell division cycle associated 8 (CDCA8) in human hepatocellular carcinoma (HCC) and its poorer survival outcome. However, the functions of CDCA8 during HCC development remain unknown. Here, we demonstrate in vitro that CDCA8 silencing inhibits HCC cell growth and long-term colony formation and migration through the accumulation of the G2/M phase cell population. Conversely, CDCA8 overexpression increases the ability to undergo long-term colony formation and migration. RNA sequencing and bioinformatic analysis revealed that CDCA8 knockdown led to the same directional regulation in 50 genes (25 down- and 25 upregulated). It was affirmed based on protein levels that CDCA8 silencing downregulates the levels of cyclin B1 and p-cdc2 and explains how it could induce G2/M arrest. The same condition increased the protein levels of tumor-suppressive ATF3 and GADD34 and inactivated AKT/ß-catenin signaling, which plays an important role in cell growth and stemness, reflecting a reduction in sphere-forming capacity. Importantly, it was demonstrated that the extent of CDCA8 expression is much greater in CD133+ cancer stem cells than in CD133- cancer cells, and that CDCA8 knockdown decreases levels of CD133, p-Akt and ß-catenin and increases levels of ATF3 and GADD34 in the CD133+ cancer stem cell (CSC) population. These molecular changes led to the inhibition of cell growth and sphere formation in the CD133+ cell population. Targeting CDCA8 also effectively suppressed tumor growth in a murine xenograft model, showing consistent molecular alterations in tumors injected with CDCA8siRNA. Taken together, these findings indicate that silencing CDCA8 suppresses HCC growth and stemness via restoring the ATF3 tumor suppressor and inactivating oncogenic AKT/ß-catenin signaling, and that targeting CDCA8 may be the next molecular strategy for both primary HCC treatment and the prevention of metastasis or recurrence.

15.
J Am Chem Soc ; 143(12): 4758-4765, 2021 03 31.
Article in English | MEDLINE | ID: mdl-33705125

ABSTRACT

Intracellular protein delivery enables selective regulation of cellular metabolism, signaling, and development through introduction of defined protein quantities into the cell. Most applications require that the delivered protein has access to the cytosol, either for protein activity or as a gateway to other organelles such as the nucleus. The vast majority of delivery vehicles employ an endosomal pathway however, and efficient release of entrapped protein cargo from the endosome remains a challenge. Recent research has made significant advances toward efficient cytosolic delivery of proteins using polymers, but the influence of polymer architecture on protein delivery is yet to be investigated. Here, we developed a family of dendronized polymers that enable systematic alterations of charge density and structure. We demonstrate that while modulation of surface functionality has a significant effect on overall delivery efficiency, the endosomal release rate can be highly regulated by manipulating polymer architecture. Notably, we show that large, multivalent structures cause slower sustained release, while rigid spherical structures result in rapid burst release.


Subject(s)
Cytosol/metabolism , Polymers/chemistry , Protein Engineering , Proteins/metabolism , Animals , Cell Line , Cytosol/chemistry , Humans , Mice , Molecular Structure , Polymers/metabolism , Proteins/chemistry
16.
Trends Pharmacol Sci ; 41(10): 743-754, 2020 10.
Article in English | MEDLINE | ID: mdl-32891429

ABSTRACT

Protein-based therapeutics have unique therapeutic potential due to their specificity, potency, and low toxicity. The vast majority of intracellular applications of proteins require access to the cytosol. Direct entry to the cytosol is challenging due to the impermeability of the cell membrane to proteins. As a result, multiple strategies have focused on endocytic uptake of proteins. Endosomally entrapped cargo, however, can have very low escape efficiency, with protein degradation occurring in acidic endolysosomal compartments. In this review, we briefly discuss endosomal escape strategies and review the strategy of cell membrane fusion, a recent strategy for direct delivery of proteins into the cell cytoplasm.


Subject(s)
Drug Delivery Systems , Proteins , Cell Membrane , Cytosol , Endosomes
17.
Adv Drug Deliv Rev ; 156: 188-213, 2020.
Article in English | MEDLINE | ID: mdl-32610061

ABSTRACT

Inorganic nanoparticles provide multipurpose platforms for a broad range of delivery applications. Intrinsic nanoscopic properties provide access to unique magnetic and optical properties. Equally importantly, the structural and functional diversity of gold, silica, iron oxide, and lanthanide-based nanocarriers provide unrivalled control of nanostructural properties for effective transport of therapeutic cargos, overcoming biobarriers on the cellular and organismal level. Taken together, inorganic nanoparticles provide a key addition to the arsenal of delivery vectors for fighting disease and improving human health.


Subject(s)
Drug Delivery Systems , Nanoparticles/administration & dosage , Nucleic Acids/administration & dosage , Proteins/administration & dosage , Animals , Ferric Compounds/administration & dosage , Gold/administration & dosage , Humans , Lanthanoid Series Elements/administration & dosage , Silicon Dioxide/administration & dosage
18.
ACS Appl Mater Interfaces ; 12(13): 15823-15829, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32150373

ABSTRACT

Intracellular delivery is essential to therapeutic applications such as genome engineering and disease diagnosis. Current methods lack simple, noninvasive strategies and are often hindered by long incubation time or high toxicity. Hydrodynamic approaches offer rapid and controllable delivery of small molecules, but thus far have not been demonstrated for delivering functional proteins. In this work, we developed a robust hydrodynamic approach based on gigahertz (GHz) acoustics to achieve rapid and noninvasive cytosolic delivery of biologically active proteins. With this method, GHz-based acoustic devices trigger oscillations through a liquid medium (acoustic streaming), generating shear stress on the cell membrane and inducing transient nanoporation. This mechanical effect enhances membrane permeability and enables cytosolic access to cationic proteins without disturbing their bioactivity. We evaluated the versatility of this approach through the delivery of cationic fluorescent proteins to a range of cell lines, all of which displayed equally efficient delivery speed (≤20 min). Delivery of multiple enzymatically active proteins with functionality related to apoptosis or genetic recombination further demonstrated the relevance of this method.


Subject(s)
Cytosol/metabolism , Proteins/metabolism , Radio Waves , Acoustics , Animals , Cell Line , Cell Survival , Cytochromes c/chemistry , Cytochromes c/metabolism , Granzymes/chemistry , Granzymes/metabolism , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/metabolism , Humans , Ions/chemistry , Mice , Proteins/chemistry
19.
J Am Chem Soc ; 142(9): 4349-4355, 2020 03 04.
Article in English | MEDLINE | ID: mdl-32049533

ABSTRACT

Nanocarrier-mediated protein delivery is a promising strategy for fundamental research and therapeutic applications. However, the efficacy of the current platforms for delivery into cells is limited by endosomal entrapment of delivered protein cargo with concomitantly inefficient access to the cytosol and other organelles, including the nucleus. We report here a robust, versatile polymeric-protein nanocomposite (PPNC) platform capable of efficient (≥90%) delivery of proteins to the cytosol. We synthesized a library of guanidinium-functionalized poly(oxanorborneneimide) (PONI) homopolymers with varying molecular weights to stabilize and deliver engineered proteins featuring terminal oligoglutamate "E-tags". The polymers were screened for cytosolic delivery efficiency using imaging flow cytometry with cytosolic delivery validated using confocal microscopy and activity of the delivered proteins demonstrated through functional assays. These studies indicate that the PPNC platform provides highly effective and tunable cytosolic delivery over a wide range of formulations, making them robust agents for therapeutic protein delivery.


Subject(s)
Drug Carriers/metabolism , Integrases/metabolism , Luminescent Proteins/metabolism , Polyglutamic Acid/metabolism , Polymers/metabolism , Drug Carriers/chemical synthesis , Guanidines/chemical synthesis , Guanidines/metabolism , HEK293 Cells , HeLa Cells , Humans , Imides/chemical synthesis , Imides/metabolism , Nanocomposites/chemistry , Polymers/chemical synthesis , Protein Engineering , Red Fluorescent Protein
20.
Theranostics ; 9(11): 3280-3292, 2019.
Article in English | MEDLINE | ID: mdl-31244954

ABSTRACT

Protein delivery into cells is a potentially transformative tool for treating "undruggable" targets in diseases associated with protein deficiencies or mutations. The vast majority of these targets are accessed via the cytosol, a challenging prospect for proteins with therapeutic and diagnostic relevance. In this review we will present promising non-viral approaches for intracellular and ultimately cytosolic delivery of proteins using nanocarriers. We will also discuss the mechanistic properties that govern the efficacy of nanocarrier-mediated protein delivery, applications of nanomaterials, and key challenges and opportunities in the use of nanocarriers for intracellular protein delivery.


Subject(s)
Cytosol/metabolism , Drug Carriers/metabolism , Drug Delivery Systems/methods , Endocytosis , Nanoparticles/metabolism , Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...