Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20.409
Filter
1.
Nat Commun ; 15(1): 3857, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38719843

ABSTRACT

Systematical and critical learning from industrial catalysis will bring inspiration for emerging nanocatalytic medicine, but the relevant knowledge is quite limited so far. In this review, we briefly summarize representative catalytic reactions and corresponding catalysts in industry, and then distinguish the similarities and differences in catalytic reactions between industrial and medical applications in support of critical learning, deep understanding, and rational designing of appropriate catalysts and catalytic reactions for various medical applications. Finally, we summarize/outlook the present and potential translation from industrial catalysis to nanocatalytic medicine. This review is expected to display a clear picture of nanocatalytic medicine evolution.


Subject(s)
Nanomedicine , Catalysis , Humans , Nanomedicine/methods , Industry , Nanotechnology/methods
2.
Nat Commun ; 15(1): 3781, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38710688

ABSTRACT

Taking inspiration from natural systems, in which molecular switches are ubiquitous in the biochemistry regulatory network, we aim to design and construct synthetic molecular switches driven by DNA-modifying enzymes, such as DNA polymerase and nicking endonuclease. The enzymatic treatments on our synthetic DNA constructs controllably switch ON or OFF the sticky end cohesion and in turn cascade to the structural association or disassociation. Here we showcase the concept in multiple DNA nanostructure systems with robust assembly/disassembly performance. The switch mechanisms are first illustrated in minimalist systems with a few DNA strands. Then the ON/OFF switches are realized in complex DNA lattice and origami systems with designated morphological changes responsive to the specific enzymatic treatments.


Subject(s)
DNA-Directed DNA Polymerase , DNA , Nanostructures , DNA/chemistry , DNA/metabolism , Nanostructures/chemistry , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/chemistry , Nucleic Acid Conformation , Deoxyribonuclease I/metabolism , Deoxyribonuclease I/chemistry , Nanotechnology/methods
3.
Sheng Wu Gong Cheng Xue Bao ; 40(5): 1323-1337, 2024 May 25.
Article in Chinese | MEDLINE | ID: mdl-38783800

ABSTRACT

Nanotechnology has attracted increasing attention in the field of medical applications due to its significant potential for development. However, one major challenge that has emerged with nanoparticles is their tendency to activate the host immune clearance system, which hampers the achievement of desired therapeutic outcomes and may lead to harmful side effects. In recent years, membrane-coated nanoparticles have emerged as a promising solution, demonstrating their effectiveness in evading immune system clearance. These innovative nanoparticles inherit essential biological attributes from natural cell membranes, such as anchoring proteins and antigens. Consequently, membrane-coated nanoparticles exhibit unique capabilities such as immune evasion, prolonged circulation, targeted drug release, and immune modulation, substantially enhancing their versatility and prospects within the realm of biomedical applications. This review provides a comprehensive overview of the current applications of cell membrane-coated nanoparticles in disease therapy, highlighting their immense potential in this rapidly evolving platform. Additionally, the review outlines the promising prospects of this technology in disease therapy.


Subject(s)
Cell Membrane , Nanoparticles , Nanoparticles/chemistry , Humans , Cell Membrane/metabolism , Drug Delivery Systems , Animals , Neoplasms/therapy , Nanotechnology/methods
4.
Sheng Wu Gong Cheng Xue Bao ; 40(5): 1352-1364, 2024 May 25.
Article in Chinese | MEDLINE | ID: mdl-38783802

ABSTRACT

In recent years, nanoscale detection has played an increasingly important role in the research on viruses, exosomes, small bacteria, and organelles. The small size and complex biological natures of these particles, with the smallest known virus particle measuring only 17 nm in diameter and exosomes ranging from 30 nm to 150 nm in size, pose challenges to the classical large-scale (typically micron-scale) characterization methods, which has become a major obstacle in the research. The emergence of nanoscale detection and analysis technologies has filled the gap of optical microscopy, a conventional technique in this field. These technologies enable the sensitive and robust detection of objects that exceed the lower limit of optical detection, revealing the molecular composition and biological roles simultaneously. Currently, several commercialized instruments based on nanotechnology have emerged, providing complete single-particle detection solutions and achieving unique functionality based on their respective technological advantages. However, it is inevitable that these technologies have limitations in terms of application and detection capabilities, as they continue to evolve. This paper offers a thorough overview of the principles, advantages, limitations, and future development trends of several mainstream commercial instruments, aiming to serve researchers in selecting and utilizing these technologies.


Subject(s)
Nanoparticles , Nanotechnology , Nanoparticles/chemistry , Nanotechnology/methods , Exosomes , Viruses/isolation & purification , Particle Size
5.
Biomed Mater ; 19(4)2024 May 23.
Article in English | MEDLINE | ID: mdl-38697209

ABSTRACT

In the field of medicine, we often brave the unknown like interstellar explorers, especially when confronting the formidable opponent of hepatocellular carcinoma (HCC). The global burden of HCC remains significant, with suboptimal treatment outcomes necessitating the urgent development of novel drugs and treatments. While various treatments for liver cancer, such as immunotherapy and targeted therapy, have emerged in recent years, improving their transport and therapeutic efficiency, controlling their targeting and release, and mitigating their adverse effects remains challenging. However, just as we grope through the darkness, a glimmer of light emerges-nanotechnology. Recently, nanotechnology has attracted attention because it can increase the local drug concentration in tumors, reduce systemic toxicity, and has the potential to enhance the effectiveness of precision therapy for HCC. However, there are also some challenges hindering the clinical translation of drug-loaded nanoparticles (NPs). Just as interstellar explorers must overcome interstellar dust, we too must overcome various obstacles. In future researches, the design and development of nanodelivery systems for novel drugs treating HCC should be the first attention. Moreover, researchers should focus on the active targeting design of various NPs. The combination of the interventional therapies and drug-loaded NPs will greatly advance the process of precision HCC therapy.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Nanoparticles , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Liver Neoplasms/therapy , Humans , Nanoparticles/chemistry , Animals , Drug Delivery Systems , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Nanotechnology/methods , Nanomedicine/methods , Immunotherapy/methods , Drug Carriers/chemistry
6.
Science ; 384(6697): 776-781, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38753798

ABSTRACT

Sophisticated statistical mechanics approaches and human intuition have demonstrated the possibility of self-assembling complex lattices or finite-size constructs. However, attempts so far have mostly only been successful in silico and often fail in experiment because of unpredicted traps associated with kinetic slowing down (gelation, glass transition) and competing ordered structures. Theoretical predictions also face the difficulty of encoding the desired interparticle interaction potential with the experimentally available nano- and micrometer-sized particles. To overcome these issues, we combine SAT assembly (a patchy-particle interaction design algorithm based on constrained optimization) with coarse-grained simulations of DNA nanotechnology to experimentally realize trap-free self-assembly pathways. We use this approach to assemble a pyrochlore three-dimensional lattice, coveted for its promise in the construction of optical metamaterials, and characterize it with small-angle x-ray scattering and scanning electron microscopy visualization.


Subject(s)
Algorithms , DNA , Nanotechnology , DNA/chemistry , Nanotechnology/methods , Scattering, Small Angle , X-Ray Diffraction , Nanostructures/chemistry , Nucleic Acid Conformation , Microscopy, Electron, Scanning
7.
J Am Chem Soc ; 146(19): 12919-12924, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38691627

ABSTRACT

RNA is a key biochemical marker, yet its chemical instability and complex secondary structure hamper its integration into DNA nanotechnology-based sensing platforms. Relying on the denaturation of the native RNA structure using urea, we show that restructured DNA/RNA hybrids can readily be prepared at room temperature. Using solid-state nanopore sensing, we demonstrate that the structures of our DNA/RNA hybrids conform to the design at the single-molecule level. Employing this chemical annealing procedure, we mitigate RNA self-cleavage, enabling the direct detection of restructured RNA molecules for biosensing applications.


Subject(s)
DNA , Nanopores , RNA , RNA/chemistry , RNA/analysis , DNA/chemistry , Biosensing Techniques/methods , Nucleic Acid Conformation , Nucleic Acid Hybridization , Nanotechnology/methods , Urea/chemistry
8.
Int J Mol Sci ; 25(9)2024 May 02.
Article in English | MEDLINE | ID: mdl-38732200

ABSTRACT

We are living in an era of advanced nanoscience and nanotechnology. Numerous nanomaterials, culminating in nanorobots, have demonstrated ingenious applications in biomedicine, including breast cancer (BC) nano-theranostics. To solve the complicated problem of BC heterogeneity, non-targeted drug distribution, invasive diagnostics or surgery, resistance to classic onco-therapies and real-time monitoring of tumors, nanorobots are designed to perform multiple tasks at a small scale, even at the organelles or molecular level. Over the last few years, most nanorobots have been bioengineered as biomimetic and biocompatible nano(bio)structures, resembling different organisms and cells, such as urchin, spider, octopus, fish, spermatozoon, flagellar bacterium or helicoidal cyanobacterium. In this review, readers will be able to deepen their knowledge of the structure, behavior and role of several types of nanorobots, among other nanomaterials, in BC theranostics. We summarized here the characteristics of many functionalized nanodevices designed to counteract the main neoplastic hallmark features of BC, from sustaining proliferation and evading anti-growth signaling and resisting programmed cell death to inducing angiogenesis, activating invasion and metastasis, preventing genomic instability, avoiding immune destruction and deregulating autophagy. Most of these nanorobots function as targeted and self-propelled smart nano-carriers or nano-drug delivery systems (nano-DDSs), enhancing the efficiency and safety of chemo-, radio- or photodynamic therapy, or the current imagistic techniques used in BC diagnosis. Most of these nanorobots have been tested in vitro, using various BC cell lines, as well as in vivo, mainly based on mice models. We are still waiting for nanorobots that are low-cost, as well as for a wider transition of these favorable effects from laboratory to clinical practice.


Subject(s)
Breast Neoplasms , Nanotechnology , Humans , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Breast Neoplasms/diagnosis , Female , Nanotechnology/methods , Animals , Nanostructures/chemistry , Nanostructures/therapeutic use , Robotics/methods , Theranostic Nanomedicine/methods , Drug Delivery Systems/methods , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology
9.
Sensors (Basel) ; 24(9)2024 May 01.
Article in English | MEDLINE | ID: mdl-38733011

ABSTRACT

Demand is strong for sensitive, reliable, and cost-effective diagnostic tools for cancer detection. Accordingly, bead-based biosensors have emerged in recent years as promising diagnostic platforms based on wide-ranging cancer biomarkers owing to the versatility, high sensitivity, and flexibility to perform the multiplexing of beads. This comprehensive review highlights recent trends and innovations in the development of bead-based biosensors for cancer-biomarker detection. We introduce various types of bead-based biosensors such as optical, electrochemical, and magnetic biosensors, along with their respective advantages and limitations. Moreover, the review summarizes the latest advancements, including fabrication techniques, signal-amplification strategies, and integration with microfluidics and nanotechnology. Additionally, the challenges and future perspectives in the field of bead-based biosensors for cancer-biomarker detection are discussed. Understanding these innovations in bead-based biosensors can greatly contribute to improvements in cancer diagnostics, thereby facilitating early detection and personalized treatments.


Subject(s)
Biomarkers, Tumor , Biosensing Techniques , Neoplasms , Biosensing Techniques/methods , Biosensing Techniques/instrumentation , Humans , Neoplasms/diagnosis , Biomarkers, Tumor/analysis , Electrochemical Techniques/methods , Nanotechnology/trends , Nanotechnology/methods , Nanotechnology/instrumentation , Microfluidics/methods , Microfluidics/instrumentation , Microfluidics/trends
10.
Ann Med ; 56(1): 2352022, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38753584

ABSTRACT

Uric acid (UA) levels in blood serum have been associated with hypertension, indicating a potential causal relationship between high serum UA levels and the progression of hypertension. Therefore, the reduction of serum UA level is considered a potential strategy for lowering and mitigating blood pressure. If an individual is at risk of developing or already manifesting elevated blood pressure, this intervention could be an integral part of a comprehensive treatment plan. By addressing hyperuricaemia, practitioners may subsidize the optimization of blood pressure regulation, which illustrates the importance of addressing UA levels as a valuable strategy within the broader context of hypertension management. In this analysis, we outlined the operational principles of effective xanthine oxidase inhibitors for the treatment of hyperuricaemia and hypertension, along with an exploration of the contribution of nanotechnology to this field.


Subject(s)
Hypertension , Hyperuricemia , Uric Acid , Xanthine Oxidase , Humans , Hyperuricemia/drug therapy , Hypertension/drug therapy , Uric Acid/blood , Xanthine Oxidase/antagonists & inhibitors , Blood Pressure/drug effects , Nanotechnology/methods , Antihypertensive Agents/therapeutic use
11.
Methods Mol Biol ; 2800: 35-53, 2024.
Article in English | MEDLINE | ID: mdl-38709476

ABSTRACT

Clustering of type II tumor necrosis factor (TNF) receptors (TNFRs) is essential for their activation, yet currently available drugs fail to activate signaling. Some strategies aim to cluster TNFR by using multivalent streptavidin or scaffolds based on dextran or graphene. However, these strategies do not allow for control of the valency or spatial organization of the ligands, and consequently control of the TNFR activation is not optimal. DNA origami nanostructures allow nanometer-precise control of the spatial organization of molecules and complexes, with defined spacing, number and valency. Here, we demonstrate the design and characterization of a DNA origami nanostructure that can be decorated with engineered single-chain TNF-related apoptosis-inducing ligand (SC-TRAIL) complexes, which show increased cell killing compared to SC-TRAIL alone on Jurkat cells. The information in this chapter can be used as a basis to decorate DNA origami nanostructures with various proteins, complexes, or other biomolecules.


Subject(s)
DNA , Nanostructures , Nanostructures/chemistry , Humans , Jurkat Cells , DNA/chemistry , DNA/metabolism , TNF-Related Apoptosis-Inducing Ligand/chemistry , TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor/chemistry , Nanotechnology/methods , Nucleic Acid Conformation
12.
Methods Mol Biol ; 2800: 103-113, 2024.
Article in English | MEDLINE | ID: mdl-38709481

ABSTRACT

The spatial resolution of conventional light microscopy is restricted by the diffraction limit to hundreds of nanometers. Super-resolution microscopy enables single digit nanometer resolution by circumventing the diffraction limit of conventional light microscopy. DNA point accumulation for imaging in nanoscale topography (DNA-PAINT) belongs to the family of single-molecule localization super-resolution approaches. Unique features of DNA-PAINT are that it allows for sub-nanometer resolution, spectrally unlimited multiplexing, proximity detection, and quantitative counting of target molecules. Here, we describe prerequisites for efficient DNA-PAINT microscopy.


Subject(s)
DNA , Single Molecule Imaging , DNA/chemistry , Single Molecule Imaging/methods , Microscopy, Fluorescence/methods , Proteins/chemistry , Nanotechnology/methods
13.
Int J Mol Sci ; 25(10)2024 May 19.
Article in English | MEDLINE | ID: mdl-38791582

ABSTRACT

A novel nanotechnology-based drug delivery system (DDS) targeted at pancreatic cancer cells was developed, characterized, and tested. The system consisted of liposomes as carriers, an anticancer drug (paclitaxel) as a chemotherapeutic agent, and a modified synthetic somatostatin analog, 5-pentacarbonyl-octreotide, a ligand for somatostatin receptor 2 (SSTR2), as a targeting moiety for pancreatic cancer. The cellular internalization, cytotoxicity, and antitumor activity of the DDS were tested in vitro using human pancreatic ductal adenocarcinoma (PDAC) cells with different expressions of the targeted SSTR2 receptors, and in vivo on immunodeficient mice bearing human PDAC xenografts. The targeted drug delivery system containing paclitaxel exhibited significantly enhanced cytotoxicity compared to non-targeted DDS, and this efficacy was directly related to the levels of SSTR2 expression. It was found that octreotide-targeted DDS proved exceptionally effective in suppressing the growth of PDAC tumors. This study underscores the potential of octreotide-targeted liposomal delivery systems to enhance the therapeutic outcomes for PDAC compared with non-targeted liposomal DDS and Paclitaxel-Cremophor® EL, suggesting a promising avenue for future cancer therapy innovations.


Subject(s)
Drug Delivery Systems , Liposomes , Octreotide , Paclitaxel , Pancreatic Neoplasms , Receptors, Somatostatin , Xenograft Model Antitumor Assays , Animals , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Receptors, Somatostatin/metabolism , Mice , Cell Line, Tumor , Paclitaxel/pharmacology , Paclitaxel/administration & dosage , Paclitaxel/therapeutic use , Liposomes/chemistry , Drug Delivery Systems/methods , Octreotide/administration & dosage , Octreotide/pharmacology , Somatostatin/analogs & derivatives , Nanotechnology/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology
14.
Int J Mol Sci ; 25(10)2024 May 19.
Article in English | MEDLINE | ID: mdl-38791578

ABSTRACT

Nanoparticles and nanomaterials are important, because they are potentially applicable to energy, storage, bioimaging, biosensors, catalysts, nanomedicine, batteries, solar energy, bioenergy, and so on (Figure 1) [...].


Subject(s)
Nanostructures , Nanostructures/chemistry , Nanomedicine/methods , Biosensing Techniques/methods , Nanotechnology/methods , Humans , Solar Energy , Nanoparticles/chemistry
15.
Molecules ; 29(10)2024 May 11.
Article in English | MEDLINE | ID: mdl-38792122

ABSTRACT

The eye's complex anatomical structures present formidable barriers to effective drug delivery across a range of ocular diseases, from anterior to posterior segment pathologies. Emerging as a promising solution to these challenges, nanotechnology-based platforms-including but not limited to liposomes, dendrimers, and micelles-have shown the potential to revolutionize ophthalmic therapeutics. These nanocarriers enhance drug bioavailability, increase residence time in targeted ocular tissues, and offer precise, localized delivery, minimizing systemic side effects. Focusing on pediatric ophthalmology, particularly on retinoblastoma, this review delves into the recent advancements in functionalized nanosystems for drug delivery. Covering the literature from 2017 to 2023, it comprehensively examines these nanocarriers' potential impact on transforming the treatment landscape for retinoblastoma. The review highlights the critical role of these platforms in overcoming the unique pediatric eye barriers, thus enhancing treatment efficacy. It underscores the necessity for ongoing research to realize the full clinical potential of these innovative drug delivery systems in pediatric ophthalmology.


Subject(s)
Drug Delivery Systems , Retinoblastoma , Retinoblastoma/drug therapy , Humans , Drug Carriers/chemistry , Child , Nanoparticles/chemistry , Micelles , Liposomes/chemistry , Dendrimers/chemistry , Retinal Neoplasms/drug therapy , Administration, Ophthalmic , Nanotechnology/methods
16.
Nat Commun ; 15(1): 4403, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38782907

ABSTRACT

Controlled manipulation of cultured cells by delivery of exogenous macromolecules is a cornerstone of experimental biology. Here we describe a platform that uses nanopipettes to deliver defined numbers of macromolecules into cultured cell lines and primary cells at single molecule resolution. In the nanoinjection platform, the nanopipette is used as both a scanning ion conductance microscope (SICM) probe and an injection probe. The SICM is used to position the nanopipette above the cell surface before the nanopipette is inserted into the cell into a defined location and to a predefined depth. We demonstrate that the nanoinjection platform enables the quantitative delivery of DNA, globular proteins, and protein fibrils into cells with single molecule resolution and that delivery results in a phenotypic change in the cell that depends on the identity of the molecules introduced. Using experiments and computational modeling, we also show that macromolecular crowding in the cell increases the signal-to-noise ratio for the detection of translocation events, thus the cell itself enhances the detection of the molecules delivered.


Subject(s)
DNA , Single Molecule Imaging , Humans , Single Molecule Imaging/methods , DNA/metabolism , DNA/chemistry , Animals , Nanotechnology/methods , Proteins/metabolism , Proteins/chemistry , Macromolecular Substances/metabolism , Macromolecular Substances/chemistry , Signal-To-Noise Ratio
17.
Int J Mol Sci ; 25(10)2024 May 14.
Article in English | MEDLINE | ID: mdl-38791398

ABSTRACT

Chimeric antigen receptor (CAR)-T cell immunotherapy represents a cutting-edge advancement in the landscape of cancer treatment. This innovative therapy has shown exceptional promise in targeting and eradicating malignant tumors, specifically leukemias and lymphomas. However, despite its groundbreaking successes, (CAR)-T cell therapy is not without its challenges. These challenges, particularly pronounced in the treatment of solid tumors, include but are not limited to, the selection of appropriate tumor antigens, managing therapy-related toxicity, overcoming T-cell exhaustion, and addressing the substantial financial costs associated with treatment. Nanomedicine, an interdisciplinary field that merges nanotechnology with medical science, offers novel strategies that could potentially address these limitations. Its application in cancer treatment has already led to significant advancements, including improved specificity in drug targeting, advancements in cancer diagnostics, enhanced imaging techniques, and strategies for long-term cancer prevention. The integration of nanomedicine with (CAR)-T cell therapy could revolutionize the treatment landscape by enhancing the delivery of genes in (CAR)-T cell engineering, reducing systemic toxicity, and alleviating the immunosuppressive effects within the tumor microenvironment. This review aims to explore how far (CAR)-T cell immunotherapy has come alone, and how nanomedicine could strengthen it into the future. Additionally, the review will examine strategies to limit the off-target effects and systemic toxicity associated with (CAR)-T cell therapy, potentially enhancing patient tolerance and treatment outcomes.


Subject(s)
Immunotherapy, Adoptive , Neoplasms , Receptors, Chimeric Antigen , Humans , Receptors, Chimeric Antigen/immunology , Neoplasms/therapy , Neoplasms/immunology , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/adverse effects , Nanotechnology/methods , Nanomedicine/methods , Animals , Tumor Microenvironment/immunology , T-Lymphocytes/immunology , Antigens, Neoplasm/immunology
18.
Biomed Mater ; 19(4)2024 May 29.
Article in English | MEDLINE | ID: mdl-38697197

ABSTRACT

Infectious diseases caused by bacterial infections are common in clinical practice. Cell membrane coating nanotechnology represents a pioneering approach for the delivery of therapeutic agents without being cleared by the immune system in the meantime. And the mechanism of infection treatment should be divided into two parts: suppression of pathogenic bacteria and suppression of excessive immune response. The membrane-coated nanoparticles exert anti-bacterial function by neutralizing exotoxins and endotoxins, and some other bacterial proteins. Inflammation, the second procedure of bacterial infection, can also be suppressed through targeting the inflamed site, neutralization of toxins, and the suppression of pro-inflammatory cytokines. And platelet membrane can affect the complement process to suppress inflammation. Membrane-coated nanoparticles treat bacterial infections through the combined action of membranes and nanoparticles, and diagnose by imaging, forming a theranostic system. Several strategies have been discovered to enhance the anti-bacterial/anti-inflammatory capability, such as synthesizing the material through electroporation, pretreating with the corresponding pathogen, membrane hybridization, or incorporating with genetic modification, lipid insertion, and click chemistry. Here we aim to provide a comprehensive overview of the current knowledge regarding the application of membrane-coated nanoparticles in preventing bacterial infections as well as addressing existing uncertainties and misconceptions.


Subject(s)
Anti-Bacterial Agents , Bacterial Infections , Cell Membrane , Nanoparticles , Humans , Cell Membrane/metabolism , Bacterial Infections/drug therapy , Nanoparticles/chemistry , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Nanomedicine/methods , Inflammation , Nanotechnology/methods , Drug Delivery Systems , Bacteria , Theranostic Nanomedicine/methods
19.
J Biochem Mol Toxicol ; 38(6): e23732, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38769657

ABSTRACT

Achieving targeted, customized, and combination therapies with clarity of the involved molecular pathways is crucial in the treatment as well as overcoming multidrug resistance (MDR) in cancer. Nanotechnology has emerged as an innovative and promising approach to address the problem of drug resistance. Developing nano-formulation-based therapies using therapeutic agents poses a synergistic effect to overcome MDR in cancer. In this review, we aimed to highlight the important pathways involved in the progression of MDR in cancer mediated through nanotechnology-based approaches that have been employed to circumvent them in recent years. Here, we also discussed the potential use of marine metabolites to treat MDR in cancer, utilizing active drug-targeting nanomedicine-based techniques to enhance selective drug accumulation in cancer cells. The discussion also provides future insights for developing complex targeted, multistage responsive nanomedical drug delivery systems for effective cancer treatments. We propose more combinational studies and their validation for the possible marine-based nanoformulations for future development.


Subject(s)
Biological Products , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Nanotechnology , Neoplasms , Humans , Biological Products/chemistry , Biological Products/therapeutic use , Biological Products/pharmacology , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Neoplasms/drug therapy , Neoplasms/metabolism , Nanotechnology/methods , Aquatic Organisms/chemistry , Animals , Nanomedicine/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Drug Delivery Systems
20.
Int J Nanomedicine ; 19: 3641-3655, 2024.
Article in English | MEDLINE | ID: mdl-38681094

ABSTRACT

DNA can be used for precise construction of complex and flexible micro-nanostructures, including DNA origami, frame nucleic acids, and DNA hydrogels. DNA nanomaterials have good biocompatibility and can enter macrophages via scavenger receptor-mediated endocytosis. DNA nanomaterials can be uniquely and flexibly designed to ensure efficient uptake by macrophages, which represents a novel strategy to regulate macrophage function. With the development of nanotechnology, major advances have been made in the design and manufacturing of DNA nanomaterials for clinical therapy. In diseases accompanied by macrophage disturbances including tumor, infectious diseases, arthritis, fibrosis, acute lung injury, and atherosclerosis, DNA nanomaterials received considerable attention as potential treatments. However, we lack sufficient information to guarantee precise targeting of macrophages by DNA nanomaterials, which precludes their therapeutic applications. In this review, we summarize recent studies of macrophage-targeting DNA nanomaterials and discuss the limitations and challenges of this approach with regard to its potential use as a biological therapy.


Subject(s)
DNA , Macrophages , Nanostructures , Humans , Nanostructures/chemistry , DNA/chemistry , Macrophages/drug effects , Animals , Biological Therapy/methods , Nanotechnology/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...