Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters










Publication year range
1.
Nanoscale ; 16(29): 14033-14056, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38990143

ABSTRACT

In recent years, there has been a growing interest in multifunctional theranostic agents capable of delivering therapeutic payloads while facilitating simultaneous diagnostic imaging of diseased sites. This approach offers a comprehensive strategy particularly valuable in dynamically evolving diseases like cancer, where combining therapy and diagnostics provides crucial insights for treatment planning. Nanoscale platforms, specifically nanogels, have emerged as promising candidates due to their stability, tunability, and multifunctionality as carriers. As a well-studied subgroup of soft polymeric nanoparticles, nanogels exhibit inherent advantages due to their size and chemical compositions, allowing for passive and active targeting of diseased tissues. Moreover, nanogels loaded with therapeutic and diagnostic agents can be designed to respond to specific stimuli at the disease site, enhancing their efficacy and specificity. This capability enables fine-tuning of theranostic platforms, garnering significant clinical interest as they can be tailored for personalized treatments. The ability to monitor tumor progression in response to treatment facilitates the adaptation of therapies according to individual patient responses, highlighting the importance of designing theranostic platforms to guide clinicians in making informed treatment decisions. Consequently, the integration of therapy and diagnostics using theranostic platforms continues to advance, offering intelligent solutions to address the challenges of complex diseases such as cancer. In this context, nanogels capable of delivering therapeutic payloads and simultaneously armed with diagnostic modalities have emerged as an attractive theranostic platform. This review focuses on advances made toward the fabrication and utilization of theranostic nanogels by highlighting examples from recent literature where their performances through a combination of therapeutic agents and imaging methods have been evaluated.


Subject(s)
Nanogels , Neoplasms , Theranostic Nanomedicine , Humans , Nanogels/chemistry , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Animals , Diagnostic Imaging/methods , Drug Carriers/chemistry , Drug Delivery Systems , Multifunctional Nanoparticles/chemistry
2.
J Nanobiotechnology ; 22(1): 328, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858780

ABSTRACT

Breast cancer bone metastasis is a terminal-stage disease and is typically treated with radiotherapy and chemotherapy, which causes severe side effects and limited effectiveness. To improve this, Sonodynamic therapy may be a more safe and effective approach in the future. Bacterial outer membrane vesicles (OMV) have excellent immune-regulating properties, including modulating macrophage polarization, promoting DC cell maturation, and enhancing anti-tumor effects. Combining OMV with Sonodynamic therapy can result in synergetic anti-tumor effects. Therefore, we constructed multifunctional nanoparticles for treating breast cancer bone metastasis. We fused breast cancer cell membranes and bacterial outer membrane vesicles to form a hybrid membrane (HM) and then encapsulated IR780-loaded PLGA with HM to produce the nanoparticles, IR780@PLGA@HM, which had tumor targeting, immune regulating, and Sonodynamic abilities. Experiments showed that the IR780@PLGA@HM nanoparticles had good biocompatibility, effectively targeted to 4T1 tumors, promoted macrophage type I polarization and DC cells activation, strengthened anti-tumor inflammatory factors expression, and presented the ability to effectively kill tumors both in vitro and in vivo, which showed a promising therapeutic effect on breast cancer bone metastasis. Therefore, the nanoparticles we constructed provided a new strategy for effectively treating breast cancer bone metastasis.


Subject(s)
Bacterial Outer Membrane , Bone Neoplasms , Breast Neoplasms , Mice, Inbred BALB C , Female , Animals , Breast Neoplasms/therapy , Breast Neoplasms/pathology , Mice , Bone Neoplasms/secondary , Bone Neoplasms/therapy , Cell Line, Tumor , Ultrasonic Therapy/methods , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Humans , Nanoparticles/chemistry , Nanoparticles/therapeutic use , RAW 264.7 Cells , Cell Membrane , Multifunctional Nanoparticles/chemistry
3.
J Nanobiotechnology ; 22(1): 354, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38902775

ABSTRACT

Fundus neovascularization diseases are a series of blinding eye diseases that seriously impair vision worldwide. Currently, the means of treating these diseases in clinical practice are continuously evolving and have rapidly revolutionized treatment opinions. However, key issues such as inadequate treatment effectiveness, high rates of recurrence, and poor patient compliance still need to be urgently addressed. Multifunctional nanomedicine can specifically respond to both endogenous and exogenous microenvironments, effectively deliver drugs to specific targets and participate in activities such as biological imaging and the detection of small molecules. Nano-in-micro (NIM) delivery systems such as metal, metal oxide and up-conversion nanoparticles (NPs), quantum dots, and carbon materials, have shown certain advantages in overcoming the presence of physiological barriers within the eyeball and are widely used in the treatment of ophthalmic diseases. Few studies, however, have evaluated the efficacy of NIM delivery systems in treating fundus neovascular diseases (FNDs). The present study describes the main clinical treatment strategies and the adverse events associated with the treatment of FNDs with NIM delivery systems and summarizes the anatomical obstacles that must be overcome. In this review, we wish to highlight the principle of intraocular microenvironment normalization, aiming to provide a more rational approach for designing new NIM delivery systems to treat specific FNDs.


Subject(s)
Drug Delivery Systems , Humans , Animals , Drug Delivery Systems/methods , Neovascularization, Pathologic/drug therapy , Fundus Oculi , Quantum Dots/chemistry , Multifunctional Nanoparticles/chemistry , Retinal Neovascularization/drug therapy , Nanomedicine/methods , Nanoparticles/chemistry
4.
Biomacromolecules ; 25(7): 4557-4568, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38899740

ABSTRACT

Rheumatoid arthritis (RA) is a complicated chronic disorder of the immune system, featured with severe inflammatory joints, synovium hyperplasia, articular cartilage, and bone damage. In the RA microenvironment, RA-involved cells, overproduced nitric oxide (NO), and pro-inflammatory cytokines are highly interplayed and mutually reinforced, which form a vicious circle and play crucial roles in the formation and progression of RA. To comprehensively break the vicious circle and obtain the maximum benefits, we have developed neutrophil membrane-camouflaged NO scavenging nanoparticles based on an NO-responsive hyaluronic acid derivative for delivery of MTX. These multifunctional nanoparticles (NNO-NPs/MTX), by inheriting the membrane functions of the source cells, possess prolonged circulation and specific localization at the inflamed sites when administrated in the body. Remarkably, NNO-NPs/MTX can neutralize the pro-inflammatory cytokines via the outer membrane receptors, scavenge NO, and be responsively disassociated to release MTX for RA-involved cell regulation and HA for lubrication in the RA sites. In a collagen-induced arthritis mouse model, NNO-NPs/MTX exhibits a significant anti-inflammation effect and effectively alleviates the characteristic RA symptoms such as synovial hyperplasia and cartilage destruction, realizing the synergistic and boosted therapeutic outcome against intractable RA. Thus, NNO-NPs/MTX provides a promising and potent platform to integrately treat RA.


Subject(s)
Arthritis, Rheumatoid , Hyaluronic Acid , Methotrexate , Nitric Oxide , Hyaluronic Acid/chemistry , Animals , Arthritis, Rheumatoid/drug therapy , Mice , Methotrexate/pharmacology , Methotrexate/administration & dosage , Methotrexate/chemistry , Nitric Oxide/metabolism , Nanoparticles/chemistry , Humans , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Drug Delivery Systems/methods , Multifunctional Nanoparticles/chemistry , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology
5.
ACS Appl Mater Interfaces ; 16(21): 27127-27138, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38747495

ABSTRACT

The excessive depositions of ß-amyloid (Aß) and abnormal level of reactive oxygen species (ROS) are considered as the important pathogenic factors of Alzheimer's disease (AD). Strategies targeting only one of them have no obvious effects in clinic. In this study, a multifunctional nanocarrier CICe@M-K that crosses the blood-brain barrier (BBB) efficiently was developed for inhibiting Aß aggregation and scavenging ROS synchronously. Antioxidant curcumin (Cur) and photosensitizer IR780 were loaded in mesoporous silica nanomaterials (MSNs). Their surfaces were grafted with cerium oxide nanoparticles (CeO2 NPs) and a short peptide K (CKLVFFAED). Living imaging showed that CICe@M-K was mainly distributed in the brain, liver, and kidneys, indicating CICe@M-K crossed BBB efficiently and accumulated in brain. After the irradiation of 808 nm laser, Cur was continuously released. Both of Cur and the peptide K can recognize and bind to Aß through multiple interaction including π-π stacking interaction, hydrophobic interaction, and hydrogen bond, inhibiting Aß aggregation. On the other hand, Cur and CeO2 NPs cooperate to relieve the oxidative stress in the brains by scavenging ROS. In vivo assays showed that the CICe@M-K could diminish Aß depositions, alleviate oxidative stress, and improve cognitive ability of the APP/PS1 AD mouse model, which demonstrated that CICe@M-K is a potential agent for AD treatment.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Curcumin , Reactive Oxygen Species , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/chemistry , Reactive Oxygen Species/metabolism , Animals , Mice , Curcumin/chemistry , Curcumin/pharmacology , Drug Carriers/chemistry , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Cerium/chemistry , Cerium/pharmacology , Humans , Antioxidants/chemistry , Antioxidants/pharmacology , Nanoparticles/chemistry , Multifunctional Nanoparticles/chemistry , Silicon Dioxide/chemistry , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use
6.
Adv Mater ; 36(27): e2314309, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38520284

ABSTRACT

Triple negative breast cancer (TNBCs), known as an immunologically cold tumor, is difficult to completely eliminate with existing monotherapies, let alone metastasis and recurrence. It is urgent to design a rational combination of multiple therapies to programmatically reconstitute tumor microenvironment (TME) and reverse the immune "cold" into "hot" inflammatory tumors to improve the therapeutic effect. Hence, in this work, a multifunctional nanosystem (FeSH NPs) that integrates metal-polyphenol coordination complex as a photothermal agent and polyphenol, salvianolic acid B (SAB) as immunomodulator is designed and fabricated for synergistic photothermal-immunotherapy of TNBCs combined with anti-PD-L1 antibody. Guided by photothermal/photoacoustic dual-mode imaging, photothermal therapy (PTT) caused by FeSH NPs induces immunogenic cell death (ICD) under 808 nm laser irradiation. Subsequently, the loaded SAB is released with the addition of deferoxamine mesylate (DFO) to remodel TME, specifically TGF-ß inhibition and PD-L1 upregulation, and eliminate the primary tumors. The combination of PTT and TME reprogramming by FeSH NPs further synergizes with anti-PD-L1 antibody to eradicate recurrence and inhibit metastasis of TNBCs concurrently. Given the biosafety of FeSH NPs throughout the lifecycle, this work provides a protocol with high clinical translational promise for comprehensive programmed therapeutics of immunologically cold tumors TNBCs.


Subject(s)
B7-H1 Antigen , Immunotherapy , Triple Negative Breast Neoplasms , Tumor Microenvironment , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/drug therapy , Animals , Mice , Tumor Microenvironment/drug effects , Humans , Cell Line, Tumor , B7-H1 Antigen/metabolism , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Female , Photothermal Therapy/methods , Polyphenols/chemistry , Polyphenols/pharmacology , Multifunctional Nanoparticles/chemistry , Transforming Growth Factor beta/metabolism , Coordination Complexes/chemistry , Coordination Complexes/therapeutic use
7.
J Mater Chem B ; 10(4): 562-570, 2022 01 26.
Article in English | MEDLINE | ID: mdl-34982089

ABSTRACT

Atherosclerosis is a global disease with an extremely high morbidity and fatality rate, so it is necessary to develop effective treatments to reduce its impact. In this work, we successfully prepared a multifunctional drug-loaded nano-delivery system with pH-responsive, CD44-targeted, and chemical-photothermal synergistic treatment. Dendritic mesoporous silica nanoparticles capped with copper sulfide (CuS) were synthesized via an oil-water biphase stratification reaction system; these served as the carrier material and encapsulated the anticoagulant drug heparin (Hep). The pH-sensitive Schiff base bond was used as a gatekeeper and targeting agent to modify hyaluronic acid (HA) on the surface of the nanocarrier. HA coating endowed the nanocomposite with the ability to respond to pH and target CD44-positive inflammatory macrophages. Based on this multifunctional nanocomposite, we achieved precise drug delivery, controlled drug release, and chemical-photothermal synergistic treatment of atherosclerosis. The in vitro drug release results showed that the nanocarriers exhibited excellent drug-controlled release properties, and could release drugs in the weakly acidic microenvironment of atherosclerotic inflammation. Cytotoxicity and cell uptake experiments indicated that nanocarriers had low cytotoxicity against RAW 264.7 cells. Modification of HA to nanocarriers can be effectively internalized by RAW 264.7 cells stimulated by lipopolysaccharide (LPS). Combining CuS photothermal treatment with anti-atherosclerosis chemotherapy showed better effects than single treatment in vitro and in vivo. In summary, our research proved that H-CuS@DMSN-NC-HA has broad application prospects in anti-atherosclerosis.


Subject(s)
Atherosclerosis/drug therapy , Hyaluronic Acid/therapeutic use , Multifunctional Nanoparticles/chemistry , Phototherapy , Animals , Cell Survival/drug effects , Copper/chemistry , Hyaluronic Acid/chemical synthesis , Hyaluronic Acid/chemistry , Hydrogen-Ion Concentration , Materials Testing , Mice , Nanoparticles/chemistry , Particle Size , RAW 264.7 Cells , Silicon Dioxide/chemistry
8.
Angew Chem Int Ed Engl ; 60(51): 26734-26739, 2021 12 13.
Article in English | MEDLINE | ID: mdl-34624158

ABSTRACT

The digestion of pathogens inside phagosomes by immune cells occurs through a sequence of reactions including acidification and proteolysis, but how the reactions are orchestrated in the right order is unclear due to a lack of methods to simultaneously measure more than one reaction in phagosomes. Here we report a bifunctional Janus-particle probe to simultaneously monitor acidification and proteolysis in single phagosomes in live cells. Each probe consists of a pH reporter and a proteolysis reporter that are spatially separated but function concurrently. Using the Janus probes, we found the acidic pH needed to initiate and maintain proteolysis, revealing the mechanism for the sequential occurrence of both reactions during pathogen digestion. We showed how bacterium-derived lipopolysaccharides alter the acidification and proteolysis in phagosomes. This study showcases Janus-particle probes as a generally applicable tool for monitoring multiple reactions in intracellular vesicles.


Subject(s)
Multifunctional Nanoparticles/metabolism , Phagosomes/metabolism , Hydrogen-Ion Concentration , Multifunctional Nanoparticles/chemistry , Particle Size , Phagosomes/chemistry , Proteolysis , Time Factors
9.
J Med Chem ; 64(18): 13853-13872, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34517696

ABSTRACT

The development of novel therapeutic strategies for combating Alzheimer's disease (AD) is challenging but imperative. Multifunctional nanoparticles are promising tools for regulating complex pathological dysfunctions for AD treatment. Herein, we constructed multifunctional nanoparticles consisting of regadenoson (Reg), nitric oxide (NO) donor, and YC-1 in a single molecular entity that can spontaneously self-assemble into nanoparticles and load donepezil to yield Reg-nanoparticles (Reg-NPs). The Reg moiety enabled the Reg-NPs to effectively regulate tight junction-associated proteins in the blood-brain barrier, thus facilitating the permeation of donepezil through the barrier and its accumulation in the brain. Moreover, the released NO and YC-1 activated the NO/cGMP/CREB signaling pathway by stimulating soluble guanylyl cyclase and inhibiting phosphodiesterase activity, which finally reduced cytotoxicity induced by aggregated Aß in the neurons and was beneficial for synaptic plasticity and memory formation.


Subject(s)
Alzheimer Disease/drug therapy , Multifunctional Nanoparticles/chemistry , Neuroprotective Agents/pharmacology , Nitric Oxide Donors/pharmacology , Nootropic Agents/pharmacology , Signal Transduction/drug effects , Alzheimer Disease/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Donepezil/chemistry , Donepezil/pharmacology , Drug Carriers/chemistry , Drug Liberation , Humans , Indazoles/chemistry , Indazoles/pharmacology , Male , Memory/drug effects , Mice, Inbred BALB C , Mice, Nude , Neuronal Plasticity/drug effects , Neurons/drug effects , Neuroprotective Agents/chemistry , Nitric Oxide/metabolism , Nitric Oxide Donors/chemistry , Nootropic Agents/chemistry , Oxadiazoles/chemistry , Oxadiazoles/pharmacology , Purines/chemistry , Purines/pharmacology , Pyrazoles/chemistry , Pyrazoles/pharmacology , Rats, Sprague-Dawley
10.
ACS Appl Mater Interfaces ; 13(39): 46406-46420, 2021 Oct 06.
Article in English | MEDLINE | ID: mdl-34569225

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease associated with amyloid-ß (Aß) deposition, leading to neurotoxicity (oxidative stress and neuroinflammation) and gut microbiota imbalance. Resveratrol (Res) has neuroprotective properties, but its bioavailability in vivo is very low. Herein, we developed a small Res-selenium-peptide nanocomposite to enable the application of Res for eliminating Aß aggregate-induced neurotoxicity and mitigating gut microbiota disorder in aluminum chloride (AlCl3) and d-galactose(d-gal)-induced AD model mice. Res functional selenium nanoparticles (Res@SeNPs) (8 ± 0.34 nm) were prepared first, after which the surface of Res@SeNPs was decorated with a blood-brain barrier transport peptide (TGN peptide) to generate Res-selenium-peptide nanocomposites (TGN-Res@SeNPs) (14 ± 0.12 nm). Oral administration of TGN-Res@SeNPs improves cognitive disorder through (1) interacting with Aß and decreasing Aß aggregation, effectively inhibiting Aß deposition in the hippocampus; (2) decreasing Aß-induced reactive oxygen species (ROS) and increasing activity of antioxidation enzymes in PC12 cells and in vivo; (3) down-regulating Aß-induced neuroinflammation via the nuclear factor kappa B/mitogen-activated protein kinase/Akt signal pathway in BV-2 cells and in vivo; and (4) alleviating gut microbiota disorder, particularly with respect to oxidative stress and inflammatory-related bacteria such as Alistipes, Helicobacter, Rikenella, Desulfovibrio, and Faecalibaculum. Thus, we anticipate that Res-selenium-peptide nanocomposites will offer a new potential strategy for the treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Drug Carriers/chemistry , Nanocomposites/chemistry , Neuroprotective Agents/therapeutic use , Resveratrol/therapeutic use , Administration, Oral , Aluminum Chloride , Alzheimer Disease/chemically induced , Amyloid beta-Peptides/metabolism , Animals , Bacteria/drug effects , Drug Carriers/administration & dosage , Drug Carriers/toxicity , Galactose , Gastrointestinal Microbiome/drug effects , Immobilized Proteins/administration & dosage , Immobilized Proteins/chemistry , Immobilized Proteins/toxicity , Male , Memory/drug effects , Mice, Inbred ICR , Multifunctional Nanoparticles/administration & dosage , Multifunctional Nanoparticles/chemistry , Multifunctional Nanoparticles/toxicity , Nanocomposites/administration & dosage , Nanocomposites/toxicity , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemistry , Oxidative Stress/drug effects , PC12 Cells , Peptide Fragments/metabolism , Peptides/administration & dosage , Peptides/chemistry , Peptides/toxicity , Protein Multimerization/drug effects , Rats , Resveratrol/administration & dosage , Resveratrol/chemistry , Selenium/administration & dosage , Selenium/chemistry , Selenium/toxicity
11.
ACS Appl Mater Interfaces ; 13(37): 43937-43951, 2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34499462

ABSTRACT

Nanotechnology has emerged as a promising solution to permanent elimination of cancer. However, nanoparticles themselves lack specificity to tumors. Due to enhanced migration to tumors, mesenchymal stem cells (MSCs) were suggested as cell-mediated delivery vehicles of nanoparticles. In this study, we have constructed a complex composed of photoluminescent quantum dots (QDs) and a photosensitizer chlorin e6 (Ce6) to obtain multifunctional nanoparticles, combining cancer diagnostic and therapeutic properties. QDs serve as energy donors-excited QDs transfer energy to the attached Ce6 via Förster resonance energy transfer, which in turn generates reactive oxygen species. Here, the physicochemical properties of the QD-Ce6 complex and singlet oxygen generation were measured, and the stability in protein-rich media was evaluated, showing that the complex remains the most stable in protein-free medium. In vitro studies on MSC and cancer cell response to the QD-Ce6 complex revealed the complex-loaded MSCs' potential to transport theranostic nanoparticles and induce cancer cell death. In vivo studies proved the therapeutic efficacy, as the survival of tumor-bearing mice was statistically significantly increased, while tumor progression and metastases were slowed down.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Lewis Lung/diagnostic imaging , Carcinoma, Lewis Lung/drug therapy , Mesenchymal Stem Cells/metabolism , Multifunctional Nanoparticles/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/radiation effects , Cadmium Compounds/chemistry , Cadmium Compounds/metabolism , Cadmium Compounds/radiation effects , Cadmium Compounds/therapeutic use , Carcinoma, Lewis Lung/metabolism , Cell Line, Tumor , Chlorophyllides/chemistry , Chlorophyllides/metabolism , Chlorophyllides/radiation effects , Chlorophyllides/therapeutic use , Female , Humans , Light , Mice, Inbred C57BL , Multifunctional Nanoparticles/chemistry , Multifunctional Nanoparticles/metabolism , Multifunctional Nanoparticles/radiation effects , Photochemotherapy/methods , Photosensitizing Agents/chemistry , Photosensitizing Agents/metabolism , Photosensitizing Agents/radiation effects , Photosensitizing Agents/therapeutic use , Precision Medicine/methods , Quantum Dots/chemistry , Quantum Dots/metabolism , Quantum Dots/radiation effects , Quantum Dots/therapeutic use , Selenium Compounds/chemistry , Selenium Compounds/metabolism , Selenium Compounds/radiation effects , Selenium Compounds/therapeutic use , Singlet Oxygen/metabolism , Sulfides/chemistry , Sulfides/metabolism , Sulfides/radiation effects , Sulfides/therapeutic use , Zinc Compounds/chemistry , Zinc Compounds/metabolism , Zinc Compounds/radiation effects , Zinc Compounds/therapeutic use
12.
Proc Natl Acad Sci U S A ; 118(38)2021 09 21.
Article in English | MEDLINE | ID: mdl-34531322

ABSTRACT

Herein, we studied localized electroporation and gene transfection of mammalian cells using a metallodielectric hybrid micromotor that is magnetically and electrically powered. Much like nanochannel-based, local electroporation of single cells, the presented micromotor was expected to increase reversible electroporation yield, relative to standard electroporation, as only a small portion of the cell's membrane (in contact with the micromotor) is affected. In contrast to methods in which the entire membrane of all cells within the sample are electroporated, the presented micromotor can perform, via magnetic steering, localized, spatially precise electroporation of the target cells that it traps and transports. In order to minimize nonselective electrical lysis of all cells within the chamber, resulting from extended exposure to an electrical field, magnetic propulsion was used to approach the immediate vicinity of the targeted cell, after which short-duration, electric-driven propulsion was activated to enable contact with the cell, followed by electroporation. In addition to local injection of fluorescent dye molecules, we demonstrated that the micromotor can enhance the introduction of plasmids into the suspension cells because of the dielectrophoretic accumulation of the plasmids in between the Janus particle and the attached cell prior to the electroporation step. Here, we chose a different strategy involving the simultaneous operation of many micromotors that are self-propelling, without external steering, and pair with cells in an autonomic manner. The locally electroporated suspension cells that are considered to be very difficult to transfect were shown to express the transfected gene, which is of significant importance for molecular biology research.


Subject(s)
Electroporation/methods , Transfection/methods , Animals , Biological Transport , Electricity , Gene Transfer Techniques , Humans , Magnetic Phenomena , Multifunctional Nanoparticles/chemistry , Plasmids , Single-Cell Analysis
13.
Nat Commun ; 12(1): 4691, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34344869

ABSTRACT

Collective guidance of out-of-equilibrium systems without using external fields is a challenge of paramount importance in active matter, ranging from bacterial colonies to swarms of self-propelled particles. Designing strategies to guide active matter and exploiting enhanced diffusion associated to its motion will provide insights for application from sensing, drug delivery to water remediation. However, achieving directed motion without breaking detailed balance, for example by asymmetric topographical patterning, is challenging. Here we engineer a two-dimensional periodic topographical design with detailed balance in its unit cell where we observe spontaneous particle edge guidance and corner accumulation of self-propelled particles. This emergent behaviour is guaranteed by a second-order non-Hermitian skin effect, a topologically robust non-equilibrium phenomenon, that we use to dynamically break detailed balance. Our stochastic circuit model predicts, without fitting parameters, how guidance and accumulation can be controlled and enhanced by design: a device guides particles more efficiently if the topological invariant characterizing it is non-zero. Our work establishes a fruitful bridge between active and topological matter, and our design principles offer a blueprint to design devices that display spontaneous, robust and predictable guided motion and accumulation, guaranteed by out-of-equilibrium topology.


Subject(s)
Models, Theoretical , Multifunctional Nanoparticles/chemistry , Equipment Design , Lab-On-A-Chip Devices , Motion , Phase Transition , Stochastic Processes
14.
Int J Biol Macromol ; 189: 443-454, 2021 Oct 31.
Article in English | MEDLINE | ID: mdl-34425122

ABSTRACT

In this contribution, we report the fabrication of multifunctional nanoparticles with gold shell over an iron oxide nanoparticles (INPs) core. The fabricated system combines the magnetic property of INPs and the surface plasmon resonance of gold. The developed nanoparticles are coated with thiolated pectin (TPGINs), which provides stability to the nanoparticles dispersion and allows the loading of hydrophobic anticancer drugs. Curcumin (Cur) is used as the model drug and an encapsulation efficiency of approximately 80% in TPGINs is observed. Cytotoxicity study with HeLa cells shows that Cur-loaded TPGINs have better viability percent (~30%) than Cur alone (~40%) at a dose of 30 µg of TPGINs. Further, annexin V-PI assay demonstrated the enhanced anticancer activity of Cur-loaded TPGINs via induction of apoptosis. The use of TPGINs leads to a significant enhancement in generating reactive oxygen species (ROS) in HeLa cells through improved radiosensitization by gamma irradiation (0.5 Gy). TPGINs are further evaluated for imparting contrast in magnetic resonance imaging (MRI) with the r2 relaxivity in the range of 11.06-13.94 s-1 µg-1 mL when measured at 7 Tesla. These experimental results indicate the potential of TPGINs for drug delivery and MR imaging.


Subject(s)
Diagnostic Imaging , Multifunctional Nanoparticles/chemistry , Neoplasms/diagnostic imaging , Neoplasms/therapy , Pectins/chemistry , Radiation Tolerance , Cell Death/drug effects , Cell Survival/drug effects , Curcumin/pharmacology , Drug Liberation , Endocytosis/drug effects , HeLa Cells , Humans , Hydrodynamics , Kinetics , Magnetic Resonance Imaging , Multifunctional Nanoparticles/ultrastructure , Particle Size , Phantoms, Imaging , Photoelectron Spectroscopy , Reactive Oxygen Species/metabolism , Spectrophotometry, Ultraviolet , Spectroscopy, Fourier Transform Infrared , Spectrum Analysis, Raman , Staining and Labeling , Thermogravimetry
15.
ACS Appl Mater Interfaces ; 13(36): 43755-43768, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34464080

ABSTRACT

Biomaterial-associated infections are a major cause of biomaterial implant failure. To prevent the initial attachment of bacteria to the implant surface, researchers have investigated various surface modification methods. However, most of these approaches also prevent the attachment, spread, and growth of mammalian cells, resulting in tissue integration failure. Therefore, the success of biomaterial implants requires an optimal balance between tissue integration (cell adhesion to biomaterial implants) and inhibition of bacterial colonization. In this regard, we synthesize bifunctional nanomaterials by functionalizing the pores and outer surfaces of periodic mesoporous organosilica (PMO) with antibacterial tetracycline (Tet) and antibacterial and cell-adhesive bipolymer poly-d-lysine (PDL), respectively. Then, the fabricated TetPMO-PDL nanomaterials are incorporated into alginate-based hydrogels to create injectable and 3D-printable nanocomposite (NC) hydrogels (AlgL-TetPMO-PDL). These bifunctional nanomaterial and 3D-printable NC hydrogel show pH-dependent release of Tet over 7 days. They also enhance the proliferation of eukaryotic cells (fibroblasts). TetPMO-PDL is inactive in reducing Pseudomonas aeruginosa, Staphylococcus aureus, and Enterococcus faecalis biofilms. However, AlgL-TetPMO-PDL shows significant antibiofilm activity against P. aeruginosa. These results suggest that the incorporation of TetPMO-PDL into AlgL may have a synergistic effect on the inhibition of the Gram-negative bacterial (P. aeruginosa) biofilm, while this has no effect on the reduction of the Gram-positive bacterial (S. aureus and E. faecalis) biofilm.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Carriers/chemistry , Hydrogels/chemistry , Multifunctional Nanoparticles/chemistry , Tetracycline/pharmacology , Alginates/chemistry , Anti-Bacterial Agents/chemistry , Biofilms/drug effects , Cell Line , Cell Proliferation/drug effects , Drug Carriers/chemical synthesis , Drug Liberation , Enterococcus faecalis/drug effects , Enterococcus faecalis/physiology , Humans , Hydrogen-Ion Concentration , Microbial Sensitivity Tests , Nanocomposites/chemistry , Polylysine/chemistry , Porosity , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Silicon Dioxide/chemistry , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Tetracycline/chemistry
16.
J Nanobiotechnology ; 19(1): 196, 2021 Jul 02.
Article in English | MEDLINE | ID: mdl-34215269

ABSTRACT

BACKGROUND: The development of alternative anti-angiogenesis therapy for choroidal neovascularization (CNV) remains a great challenge. Nanoparticle systems have emerged as a new form of drug delivery in ocular diseases. Here, we report the construction and characterization of arginine-glycine-aspartic acid (RGD)-conjugated polyethyleneimine (PEI) as a vehicle to load antioxidant salvianolic acid A (SAA) for targeted anti-angiogenesis therapy of CNV. In this study, PEI was consecutively modified with polyethylene glycol (PEG) conjugated RGD segments, 3-(4'-hydroxyphenyl) propionic acid-Osu (HPAO), and fluorescein isothiocyanate (FI), followed by acetylation of the remaining PEI surface amines to generate the multifunctional PEI vehicle PEI.NHAc-FI-HPAO-(PEG-RGD) (for short, RGD-PEI). The formed RGD-PEI was utilized as an effective vehicle platform to load SAA. RESULTS: We showed that RGD-PEI/SAA complexes displayed desirable water dispersibility, low cytotoxicity, and sustainable release of SAA under different pH conditions. It could be specifically taken up by retinal pigment epithelium (RPE) cells which highly expressed ɑvß5 integrin receptors in vitro and selectively accumulated in CNV lesions in vivo. Moreover, the complexes displayed specific therapeutic efficacy in a mouse model of laser induced CNV, and the slow elimination of the complexes in the vitreous cavity was verified by SPECT imaging after 131I radiolabeling. The histological examinations further confirmed the biocompatibility of RGD-PEI/SAA. CONCLUSIONS: The results suggest that the designed RGD-PEI/SAA complexes may be a potential alternative anti-angiogenesis therapy for posterior ocular neovascular diseases.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Choroidal Neovascularization/drug therapy , Multifunctional Nanoparticles/chemistry , Oligopeptides/chemistry , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Animals , Caffeic Acids , Cell Line, Tumor , Choroidal Neovascularization/pathology , Disease Models, Animal , Drug Liberation , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Lactates , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Polyethyleneimine/chemistry , Proton Pump Inhibitors/chemistry , Proton Pump Inhibitors/pharmacology , Wound Healing/drug effects
17.
ACS Appl Mater Interfaces ; 13(24): 28802-28817, 2021 Jun 23.
Article in English | MEDLINE | ID: mdl-34109788

ABSTRACT

In this study, a novel class of multifunctional responsive nanoparticles is designed and fabricated as drug nanocarriers for synergetic chemo-photothermal therapy of tumors. The proposed nanoparticles are composed of a thermo-/pH-responsive poly(N-isopropylacrylamide-co-acrylic acid) (PNA) nanogel core, a polydopamine (PDA) layer for photothermal conversion, and an outer folic acid (FA) layer as a targeting agent for the folate receptors on tumor cells. The fabricated nanoparticles show good biocompatibility and outstanding photothermal conversion efficiency. The proposed nanoparticles loaded with doxorubicin (DOX) drug molecules are stable under physiological conditions with low leakage of drugs, while rapidly release drugs in environments with low pH conditions and at high temperature. The experimental results show that the drug release process is mainly governed by Fickian diffusion. In vitro cell experimental results demonstrate that the PNA-DOX@PDA-FA nanoparticles can be phagocytized by 4T1 tumor cells and release drugs in tumor cell acidic environments, and confirm that the combined chemo and photothermal therapeutic efficacy of PNA-DOX@PDA-FA nanoparticles is higher than the photothermal therapeutic efficacy or the chemotherapeutic efficacy alone. The proposed multifunctional responsive nanoparticles in this study provide a novel class of drug nanocarriers as a promising tool for synergetic chemo-photothermal therapy of tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Doxorubicin/pharmacology , Drug Carriers/chemistry , Multifunctional Nanoparticles/chemistry , Acrylamides/chemistry , Acrylamides/metabolism , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Doxorubicin/chemistry , Drug Carriers/metabolism , Drug Carriers/radiation effects , Drug Liberation , Endocytosis/physiology , Folic Acid/analogs & derivatives , Folic Acid/metabolism , Humans , Hydrogen-Ion Concentration , Indoles/chemistry , Indoles/metabolism , Indoles/radiation effects , Infrared Rays , Mice , Multifunctional Nanoparticles/metabolism , Photothermal Therapy , Polymers/chemistry , Polymers/metabolism , Polymers/radiation effects , Temperature
19.
Mol Ther ; 29(10): 2931-2948, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34023507

ABSTRACT

Checkpoint inhibitors, such as anti-PD-1/PD-L1 antibodies, have been shown to be extraordinarily effective, but their durable response rate remains low, especially in colorectal cancer (CRC). Recent studies have shown that photodynamic therapy (PDT) could effectively enhance PD-L1 blockade therapeutic effects, although the reason is still unclear. Here, we report the use of multifunctional nanoparticles (NPs) loaded with photosensitized mTHPC (mTHPC@VeC/T-RGD NPs)-mediated PDT treatment to potentiate the anti-tumor efficacy of PD-L1 blockade for CRC treatment and investigate the underlying mechanisms of PDT enhancing PD-L1 blockade therapeutic effect in this combination therapy. In this study, the mTHPC@VeC/T-RGD NPs under the 660-nm near infrared (NIR) laser could kill tumor cells by inducing apoptosis and/or necrosis and stimulating systemic immune response, which could be further promoted by the PD-L1 blockade to inhibit primary and distant tumor growth, as well as building long-term host immunological memory to prevent tumor recurrence. Furthermore, we detected that mTHPC@VeC/T-RGD NP-mediated PDT sensitizes tumors to PD-L1 blockade therapy mainly because PDT-mediated hypoxia could induce the hypoxia-inducible factor 1α (HIF-1α) signaling pathway that upregulates PD-L1 expression in CRC. Taken together, our work demonstrates that mTHPC@VeC/T-RGD NP-mediated PDT is a promising strategy that may potentiate the response rate of anti-PD-L1 checkpoint blockade immunotherapies in CRC.


Subject(s)
Colorectal Neoplasms/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immune Checkpoint Inhibitors/administration & dosage , Photochemotherapy/methods , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/genetics , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Immune Checkpoint Inhibitors/pharmacology , Mesoporphyrins/chemistry , Mesoporphyrins/pharmacology , Mice , Multifunctional Nanoparticles/administration & dosage , Multifunctional Nanoparticles/chemistry , Particle Size , Tumor Hypoxia/drug effects
20.
Biomolecules ; 11(4)2021 03 30.
Article in English | MEDLINE | ID: mdl-33808293

ABSTRACT

The global impact of cancer emphasizes the importance of developing innovative, effective and minimally invasive therapies. In the context of superficial cancers, the development of a multifunctional nanoparticle-based system and its in vitro and in vivo safety and efficacy characterization are, herein, proposed as a proof-of-concept. This multifunctional system consists of gold nanoparticles coated with hyaluronic and oleic acids, and functionalized with epidermal growth factor for greater specificity towards cutaneous melanoma cells. This nanoparticle system is activated by a near-infrared laser. The characterization of this nanoparticle system included several phases, with in vitro assays being firstly performed to assess the safety of gold nanoparticles without laser irradiation. Then, hairless immunocompromised mice were selected for a xenograft model upon inoculation of A375 human melanoma cells. Treatment with near-infrared laser irradiation for five minutes combined with in situ administration of the nanoparticles showed a tumor volume reduction of approximately 80% and, in some cases, led to the formation of several necrotic foci, observed histologically. No significant skin erythema at the irradiation zone was verified, nor other harmful effects on the excised organs. In conclusion, these assays suggest that this system is safe and shows promising results for the treatment of superficial melanoma.


Subject(s)
Low-Level Light Therapy/methods , Melanoma/therapy , Multifunctional Nanoparticles/therapeutic use , Skin Neoplasms/therapy , Animals , Cell Line, Tumor , Epidermal Growth Factor/chemistry , Gold/chemistry , Humans , Low-Level Light Therapy/adverse effects , Male , Melanoma/pathology , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice, SCID , Multifunctional Nanoparticles/chemistry , Oleic Acid/chemistry , Proof of Concept Study , Skin Neoplasms/pathology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL