Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.901
Filter
Add more filters

Publication year range
1.
Cell ; 174(2): 259-270.e11, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29937224

ABSTRACT

Many community- and hospital-acquired bacterial infections are caused by antibiotic-resistant pathogens. Methicillin-resistant Staphylococcus aureus (MRSA) predisposes humans to invasive infections that are difficult to eradicate. We designed a closed-loop gene network programming mammalian cells to autonomously detect and eliminate bacterial infections. The genetic circuit contains human Toll-like receptors as the bacterial sensor and a synthetic promoter driving reversible and adjustable expression of lysostaphin, a bacteriolytic enzyme highly lethal to S. aureus. Immunomimetic designer cells harboring this genetic circuit exhibited fast and robust sense-and-destroy kinetics against live staphylococci. When tested in a foreign-body infection model in mice, microencapsulated cell implants prevented planktonic MRSA infection and reduced MRSA biofilm formation by 91%. Notably, this system achieved a 100% cure rate of acute MRSA infections, whereas conventional vancomycin treatment failed. These results suggest that immunomimetic designer cells could offer a therapeutic approach for early detection, prevention, and cure of pathogenic infections in the post-antibiotic era.


Subject(s)
Biomimetics/methods , Methicillin-Resistant Staphylococcus aureus/physiology , Staphylococcal Infections/prevention & control , Alkaline Phosphatase/blood , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Disk Diffusion Antimicrobial Tests , Female , HEK293 Cells , Humans , Lipopolysaccharide Receptors/genetics , Lysostaphin/metabolism , Lysostaphin/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice , Mice, Inbred C57BL , Plasmids/genetics , Plasmids/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/veterinary , Toll-Like Receptor 1/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 6/genetics , Transcription Factor AP-1/metabolism
2.
Nature ; 631(8021): 549-555, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38926572

ABSTRACT

Chemical reaction networks, such as those found in metabolism and signalling pathways, enable cells to process information from their environment1,2. Current approaches to molecular information processing and computation typically pursue digital computation models and require extensive molecular-level engineering3. Despite considerable advances, these approaches have not reached the level of information processing capabilities seen in living systems. Here we report on the discovery and implementation of a chemical reservoir computer based on the formose reaction4. We demonstrate how this complex, self-organizing chemical reaction network can perform several nonlinear classification tasks in parallel, predict the dynamics of other complex systems and achieve time-series forecasting. This in chemico information processing system provides proof of principle for the emergent computational capabilities of complex chemical reaction networks, paving the way for a new class of biomimetic information processing systems.


Subject(s)
Biomimetics , Models, Chemical , Biomimetics/methods , Machine Learning
3.
Nature ; 613(7945): 682-688, 2023 01.
Article in English | MEDLINE | ID: mdl-36653452

ABSTRACT

Helical structures are ubiquitous in nature and impart unique mechanical properties and multifunctionality1. So far, synthetic architectures that mimic these natural systems have been fabricated by winding, twisting and braiding of individual filaments1-7, microfluidics8,9, self-shaping1,10-13 and printing methods14-17. However, those fabrication methods are unable to simultaneously create and pattern multimaterial, helically architected filaments with subvoxel control in arbitrary two-dimensional (2D) and three-dimensional (3D) motifs from a broad range of materials. Towards this goal, both multimaterial18-23 and rotational24 3D printing of architected filaments have recently been reported; however, the integration of these two capabilities has yet to be realized. Here we report a rotational multimaterial 3D printing (RM-3DP) platform that enables subvoxel control over the local orientation of azimuthally heterogeneous architected filaments. By continuously rotating a multimaterial nozzle with a controlled ratio of angular-to-translational velocity, we have created helical filaments with programmable helix angle, layer thickness and interfacial area between several materials within a given cylindrical voxel. Using this integrated method, we have fabricated functional artificial muscles composed of helical dielectric elastomer actuators with high fidelity and individually addressable conductive helical channels embedded within a dielectric elastomer matrix. We have also fabricated hierarchical lattices comprising architected helical struts containing stiff springs within a compliant matrix. Our additive-manufacturing platform opens new avenues to generating multifunctional architected matter in bioinspired motifs.


Subject(s)
Artificial Organs , Biomimetic Materials , Biomimetics , Elastomers/chemistry , Electric Conductivity , Printing, Three-Dimensional , Biomimetics/methods , Biomimetic Materials/chemistry , Rotation , Muscles/chemistry
4.
Nature ; 610(7931): 283-289, 2022 10.
Article in English | MEDLINE | ID: mdl-36224418

ABSTRACT

The current proliferation of mobile robots spans ecological monitoring, warehouse management and extreme environment exploration, to an individual consumer's home1-4. This expanding frontier of applications requires robots to transit multiple environments, a substantial challenge that traditional robot design strategies have not effectively addressed5,6. For example, biomimetic design-copying an animal's morphology, propulsion mechanism and gait-constitutes one approach, but it loses the benefits of engineered materials and mechanisms that can be exploited to surpass animal performance7,8. Other approaches add a unique propulsive mechanism for each environment to the same robot body, which can result in energy-inefficient designs9-11. Overall, predominant robot design strategies favour immutable structures and behaviours, resulting in systems incapable of specializing across environments12,13. Here, to achieve specialized multi-environment locomotion through terrestrial, aquatic and the in-between transition zones, we implemented 'adaptive morphogenesis', a design strategy in which adaptive robot morphology and behaviours are realized through unified structural and actuation systems. Taking inspiration from terrestrial and aquatic turtles, we built a robot that fuses traditional rigid components and soft materials to radically augment the shape of its limbs and shift its gaits for multi-environment locomotion. The interplay of gait, limb shape and the environmental medium revealed vital parameters that govern the robot's cost of transport. The results attest that adaptive morphogenesis is a powerful method to enhance the efficiency of mobile robots encountering unstructured, changing environments.


Subject(s)
Biomimetics , Environment , Equipment Design , Robotics , Animals , Biomimetics/instrumentation , Biomimetics/methods , Locomotion , Robotics/instrumentation , Robotics/methods , Turtles/physiology
5.
Proc Natl Acad Sci U S A ; 121(33): e2407971121, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39110725

ABSTRACT

Artificial neuromorphic devices can emulate dendric integration, axonal parallel transmission, along with superior energy efficiency in facilitating efficient information processing, offering enormous potential for wearable electronics. However, integrating such circuits into textiles to achieve biomimetic information perception, processing, and control motion feedback remains a formidable challenge. Here, we engineer a quasi-solid-state iontronic synapse fiber (ISF) comprising photoresponsive TiO2, ion storage Co-MoS2, and an ion transport layer. The resulting ISF achieves inherent short-term synaptic plasticity, femtojoule-range energy consumption, and the ability to transduce chemical/optical signals. Multiple ISFs are interwoven into a synthetic neural fabric, allowing the simultaneous propagation of distinct optical signals for transmitting parallel information. Importantly, IFSs with multiple input electrodes exhibit spatiotemporal information integration. As a proof of concept, a textile-based multiplexing neuromorphic sensorimotor system is constructed to connect synaptic fibers with artificial fiber muscles, enabling preneuronal sensing information integration, parallel transmission, and postneuronal information output to control the coordinated motor of fiber muscles. The proposed fiber system holds enormous promise in wearable electronics, soft robotics, and biomedical engineering.


Subject(s)
Synapses , Textiles , Synapses/physiology , Wearable Electronic Devices , Biomimetics/methods , Biomimetics/instrumentation , Humans , Neuronal Plasticity/physiology
6.
Nature ; 585(7826): 518-523, 2020 09.
Article in English | MEDLINE | ID: mdl-32968256

ABSTRACT

Current hardware approaches to biomimetic or neuromorphic artificial intelligence rely on elaborate transistor circuits to simulate biological functions. However, these can instead be more faithfully emulated by higher-order circuit elements that naturally express neuromorphic nonlinear dynamics1-4. Generating neuromorphic action potentials in a circuit element theoretically requires a minimum of third-order complexity (for example, three dynamical electrophysical processes)5, but there have been few examples of second-order neuromorphic elements, and no previous demonstration of any isolated third-order element6-8. Using both experiments and modelling, here we show how multiple electrophysical processes-including Mott transition dynamics-form a nanoscale third-order circuit element. We demonstrate simple transistorless networks of third-order elements that perform Boolean operations and find analogue solutions to a computationally hard graph-partitioning problem. This work paves a way towards very compact and densely functional neuromorphic computing primitives, and energy-efficient validation of neuroscientific models.


Subject(s)
Artificial Intelligence , Biomimetics/methods , Computer Simulation , Engineering/methods , Models, Neurological , Action Potentials , Electrodes , Electrophysiology , Logic
7.
Nature ; 569(7755): 208-214, 2019 05.
Article in English | MEDLINE | ID: mdl-31068721

ABSTRACT

Software implementations of brain-inspired computing underlie many important computational tasks, from image processing to speech recognition, artificial intelligence and deep learning applications. Yet, unlike real neural tissue, traditional computing architectures physically separate the core computing functions of memory and processing, making fast, efficient and low-energy computing difficult to achieve. To overcome such limitations, an attractive alternative is to design hardware that mimics neurons and synapses. Such hardware, when connected in networks or neuromorphic systems, processes information in a way more analogous to brains. Here we present an all-optical version of such a neurosynaptic system, capable of supervised and unsupervised learning. We exploit wavelength division multiplexing techniques to implement a scalable circuit architecture for photonic neural networks, successfully demonstrating pattern recognition directly in the optical domain. Such photonic neurosynaptic networks promise access to the high speed and high bandwidth inherent to optical systems, thus enabling the direct processing of optical telecommunication and visual data.


Subject(s)
Biomimetics/methods , Models, Neurological , Neural Networks, Computer , Pattern Recognition, Automated/methods , Photons , Supervised Machine Learning , Unsupervised Machine Learning , Action Potentials , Computer Systems , Computers , Nerve Net/cytology , Nerve Net/physiology , Neurons/cytology , Neurons/physiology , Synapses/physiology
8.
Drug Resist Updat ; 75: 101098, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38833804

ABSTRACT

Breakthroughs in actual clinical applications have begun through vaccine-based cancer immunotherapy, which uses the body's immune system, both humoral and cellular, to attack malignant cells and fight diseases. However, conventional vaccine approaches still face multiple challenges eliciting effective antigen-specific immune responses, resulting in immunotherapy resistance. In recent years, biomimetic nanovaccines have emerged as a promising alternative to conventional vaccine approaches by incorporating the natural structure of various biological entities, such as cells, viruses, and bacteria. Biomimetic nanovaccines offer the benefit of targeted antigen-presenting cell (APC) delivery, improved antigen/adjuvant loading, and biocompatibility, thereby improving the sensitivity of immunotherapy. This review presents a comprehensive overview of several kinds of biomimetic nanovaccines in anticancer immune response, including cell membrane-coated nanovaccines, self-assembling protein-based nanovaccines, extracellular vesicle-based nanovaccines, natural ligand-modified nanovaccines, artificial antigen-presenting cells-based nanovaccines and liposome-based nanovaccines. We also discuss the perspectives and challenges associated with the clinical translation of emerging biomimetic nanovaccine platforms for sensitizing cancer cells to immunotherapy.


Subject(s)
Antigen-Presenting Cells , Cancer Vaccines , Immunotherapy , Nanoparticles , Neoplasms , Humans , Neoplasms/therapy , Neoplasms/immunology , Immunotherapy/methods , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Nanoparticles/administration & dosage , Antigen-Presenting Cells/immunology , Biomimetics/methods , Biomimetic Materials/administration & dosage , Animals , Liposomes , Nanovaccines
9.
Proc Natl Acad Sci U S A ; 119(33): e2201776119, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35943987

ABSTRACT

Many natural organisms, such as fungal hyphae and plant roots, grow at their tips, enabling the generation of complex bodies composed of natural materials as well as dexterous movement and exploration. Tip growth presents an exemplary process by which materials synthesis and actuation are coupled, providing a blueprint for how growth could be realized in a synthetic system. Herein, we identify three underlying principles essential to tip-based growth of biological organisms: a fluid pressure driving force, localized polymerization for generating structure, and fluid-mediated transport of constituent materials. In this work, these evolved features inspire a synthetic materials growth process called extrusion by self-lubricated interface photopolymerization (E-SLIP), which can continuously fabricate solid profiled polymer parts with tunable mechanical properties from liquid precursors. To demonstrate the utility of E-SLIP, we create a tip-growing soft robot, outline its fundamental governing principles, and highlight its capabilities for growth at speeds up to 12 cm/min and lengths up to 1.5 m. This growing soft robot is capable of executing a range of tasks, including exploration, burrowing, and traversing tortuous paths, which highlight the potential for synthetic growth as a platform for on-demand manufacturing of infrastructure, exploration, and sensing in a variety of environments.


Subject(s)
Bioengineering , Biomimetics , Polymerization , Robotics , Agaricales/growth & development , Bioengineering/methods , Biomimetics/methods , Movement , Plant Development
10.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Article in English | MEDLINE | ID: mdl-35181608

ABSTRACT

Dynamic biomaterials excel at recapitulating the reversible interlocking and remoldable structure of the extracellular matrix (ECM), particularly in manipulating cell behaviors and adapting to tissue morphogenesis. While strategies based on dynamic chemistries have been extensively studied for ECM-mimicking dynamic biomaterials, biocompatible molecular means with biogenicity are still rare. Here, we report a nature-derived strategy for fabrication of dynamic biointerface as well as a three-dimensional (3D) hydrogel structure based on reversible receptor-ligand interaction between the glycopeptide antibiotic vancomycin and dipeptide d-Ala-d-Ala. We demonstrate the reversible regulation of multiple cell types with the dynamic biointerface and successfully implement the dynamic hydrogel as a functional antibacterial 3D scaffold to treat tissue repair. In view of the biogenicity and high applicability, this nature-derived reversible molecular strategy will bring opportunities for malleable biomaterial design with great potential in biomedicine.


Subject(s)
Extracellular Matrix/chemistry , Extracellular Matrix/physiology , Protein Engineering/methods , Alanine/chemistry , Alanine/metabolism , Biocompatible Materials/chemistry , Biomimetics/methods , Dipeptides/metabolism , Humans , Hydrogels/chemistry , Ligands , Vancomycin/chemistry , Vancomycin/metabolism
11.
Chem Soc Rev ; 53(9): 4490-4606, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38502087

ABSTRACT

Living organisms in nature have undergone continuous evolution over billions of years, resulting in the formation of high-performance fracture-resistant biomineralized tissues such as bones and teeth to fulfill mechanical and biological functions, despite the fact that most inorganic biominerals that constitute biomineralized tissues are weak and brittle. During the long-period evolution process, nature has evolved a number of highly effective and smart strategies to design chemical compositions and structures of biomineralized tissues to enable superior properties and to adapt to surrounding environments. Most biomineralized tissues have hierarchically ordered structures consisting of very small building blocks on the nanometer scale (nanoparticles, nanofibers or nanoflakes) to reduce the inherent weaknesses and brittleness of corresponding inorganic biominerals, to prevent crack initiation and propagation, and to allow high defect tolerance. The bioinspired principles derived from biomineralized tissues are indispensable for designing and constructing high-performance biomimetic materials. In recent years, a large number of high-performance biomimetic materials have been prepared based on these bioinspired principles with a large volume of literature covering this topic. Therefore, a timely and comprehensive review on this hot topic is highly important and contributes to the future development of this rapidly evolving research field. This review article aims to be comprehensive, authoritative, and critical with wide general interest to the science community, summarizing recent advances in revealing the formation processes, composition, and structures of biomineralized tissues, providing in-depth insights into guidelines derived from biomineralized tissues for the design and construction of high-performance biomimetic materials, and discussing recent progress, current research trends, key problems, future main research directions and challenges, and future perspectives in this exciting and rapidly evolving research field.


Subject(s)
Biomimetic Materials , Biomimetic Materials/chemistry , Biomimetic Materials/metabolism , Humans , Animals , Biomineralization , Bone and Bones/chemistry , Bone and Bones/metabolism , Biomimetics/methods , Tooth/chemistry
12.
Nano Lett ; 24(26): 8107-8116, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38888223

ABSTRACT

The integration of sonodynamic therapy (SDT) with cuproptosis for targeted cancer treatment epitomizes a significant advancement in oncology. Herein, we present a dual-responsive therapeutic system, "CytoNano", which combines a cationic liposome infused with copper-nitride nanoparticles and oxygen-rich perfluorocarbon (Lip@Cu3N/PFC-O2), all enveloped in a biomimetic coating of neutrophil membrane and acid-responsive carboxymethylcellulose. CytoNano leverages the cellular mimicry of neutrophils and acid-responsive materials, enabling precise targeting of tumors and their acidic microenvironment. This strategic design facilitates the targeted release of Lip@Cu3N/PFC-O2 within the tumor, enhancing cancer cell uptake and mitochondrial localization. Consequently, it amplifies the therapeutic efficacy of both Cu3N-driven SDT and cuproptosis while preserving healthy tissues. Additionally, CytoNano's ultrasound responsiveness enhances intratumoral oxygenation, overcoming physiological barriers and initiating a combined sonodynamic-cuproptotic effect that induces multiple cell death pathways. Thus, we pioneer a biomimetic approach in precise sonodynamic cuproptosis, revolutionizing cancer therapy.


Subject(s)
Mitochondria , Ultrasonic Therapy , Humans , Mitochondria/metabolism , Mitochondria/drug effects , Animals , Ultrasonic Therapy/methods , Mice , Cell Line, Tumor , Neoplasms/therapy , Neoplasms/pathology , Nanoparticles/chemistry , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Copper/chemistry , Copper/pharmacology , Liposomes/chemistry , Fluorocarbons/chemistry , Biomimetics/methods , Oxygen/chemistry
13.
J Am Chem Soc ; 146(22): 15096-15107, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38773940

ABSTRACT

For cationic nanoparticles, the spontaneous nanoparticle-protein corona formation and aggregation in biofluids can trigger unexpected biological reactions. Herein, we present a biomimetic strategy for camouflaging the cationic peptide/siRNA nanocomplex (P/Si) with single or dual proteins, which exploits the unique properties of endogenous proteins and stabilizes the cationic P/Si complex for safe and targeted delivery. An in-depth study of the P/Si protein corona (P/Si-PC) formation and protein binding was conducted. The results provided insights into the biochemical and toxicological properties of cationic nanocomplexes and the rationales for engineering biomimetic protein camouflages. Based on this, the human serum albumin (HSA) and apolipoprotein AI (Apo-AI) ranked within the top 20 abundant protein species of P/Si-PC were selected to construct biomimetic HSA-dressed P/Si (P/Si@HSA) and dual protein (HSA and Apo-AI)-dressed P/Si (P/Si@HSA_Apo), given that the dual-protein camouflage plays complementary roles in efficient delivery. A branched cationic peptide (b-HKR) was tailored for siRNA delivery, and their nanocomplexes, including the cationic P/Si and biomimetic protein-dressed P/Si, were produced by a precise microfluidic technology. The biomimetic anionic protein camouflage greatly enhanced P/Si biostability and biocompatibility, which offers a reliable strategy for overcoming the limitation of applying cationic nanoparticles in biofluids and systemic delivery.


Subject(s)
Biomimetic Materials , Nanoparticles , Peptides , RNA, Small Interfering , Serum Albumin, Human , Humans , RNA, Small Interfering/chemistry , Peptides/chemistry , Biomimetic Materials/chemistry , Nanoparticles/chemistry , Serum Albumin, Human/chemistry , Protein Engineering , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Protein Corona/chemistry , Biomimetics/methods
14.
Small ; 20(27): e2309269, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38308170

ABSTRACT

3D printing and electrospinning are versatile techniques employed to produce 3D structures, such as scaffolds and ultrathin fibers, facilitating the creation of a cellular microenvironment in vitro. These two approaches operate on distinct working principles and utilize different polymeric materials to generate the desired structure. This review provides an extensive overview of these techniques and their potential roles in biomedical applications. Despite their potential role in fabricating complex structures, each technique has its own limitations. Electrospun fibers may have ambiguous geometry, while 3D-printed constructs may exhibit poor resolution with limited mechanical complexity. Consequently, the integration of electrospinning and 3D-printing methods may be explored to maximize the benefits and overcome the individual limitations of these techniques. This review highlights recent advancements in combined techniques for generating structures with controlled porosities on the micro-nano scale, leading to improved mechanical structural integrity. Collectively, these techniques also allow the fabrication of nature-inspired structures, contributing to a paradigm shift in research and technology. Finally, the review concludes by examining the advantages, disadvantages, and future outlooks of existing technologies in addressing challenges and exploring potential opportunities.


Subject(s)
Nanofibers , Printing, Three-Dimensional , Tissue Engineering , Tissue Scaffolds , Nanofibers/chemistry , Tissue Engineering/methods , Humans , Tissue Scaffolds/chemistry , Biomimetics/methods , Regeneration , Animals , Biomimetic Materials/chemistry
15.
Small ; 20(28): e2400165, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38329189

ABSTRACT

Biomimetic tactile nervous system (BTNS) inspired by organisms has motivated extensive attention in wearable fields due to its biological similarity, low power consumption, and perception-memory integration. Though many works about planar-shape BTNS are developed, few researches could be found in the field of fibrous BTNS (FBTNS) which is superior in terms of strong flexibility, weavability, and high-density integration. Herein, a FBTNS with multimodal sensibility and memory is proposed, by fusing the fibrous poly lactic acid (PLA)/Ag/MXene/Pt artificial synapse and MXene/EMIMBF4 ionic conductive elastomer. The proposed FBTNS can successfully perceive external stimuli and generate synaptic responses. It also exhibits a short response time (23 ms) and low set power consumption (17 nW). Additionally, the proposed device demonstrates outstanding synaptic plasticity under both mechanical and electrical stimuli, which can simulate the memory function. Simultaneously, the fibrous devices are embedded into textiles to construct tactile arrays, by which biomimetic tactile perception and temporary memory functions are successfully implemented. This work demonstrates the as-prepared FBTNS can generate biomimetic synaptic signals to serve as artificial feeling signals, it is thought that it could offer a fabric electronic unit integrating with perception and memory for Human-Computer interaction, and has great potential to build lightweight and comfortable Brain-Computer interfaces.


Subject(s)
Biomimetics , Synapses , Biomimetics/methods , Synapses/physiology , Touch/physiology , Memory/physiology , Biomimetic Materials/chemistry , Humans , Polyesters/chemistry
16.
Small ; 20(24): e2311275, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38196019

ABSTRACT

Nanomaterials with biomimetic catalytic abilities have attracted significant attention. However, the stereoselectivity of natural enzymes determined by their unique configurations is difficult to imitate. In this work, a kind of chiral CuxCoyS-CuzS nanoflowers (L/D-Pen-NFs) is developed, using porous CuxCoyS nanoparticles (NPs) as stamens, CuzS sheets as petals, and chiral penicillamine as surface stabilizers. Compared to the natural laccase enzyme, L/D-Pen-NFs exhibit significant advantages in catalytic efficiency, stability against harsh environments, recyclability, and convenience in construction. Most importantly, they display high enantioselectivity toward chiral neurotransmitters, which is proved by L- and D-Pen-NFs' different catalytic efficiencies toward chiral enantiomers. L-Pen-NFs are more efficient in catalyzing the oxidation of L-epinephrine and L-dopamine compared with D-Pen-NFs. However, their catalytic efficiency in oxidizing L-norepinephrine and L-DOPA is lower than that of D-Pen-NFs. The reason for the difference in catalytic efficiency is the distinct binding affinities between CuxCoyS-CuzS nano-enantiomers and chiral molecules. This work can spur the development of chiral nanostructures with biomimetic functions.


Subject(s)
Copper , Catalysis , Copper/chemistry , Stereoisomerism , Nanostructures/chemistry , Biomimetics/methods , Oxidation-Reduction , Laccase/chemistry , Laccase/metabolism
17.
Small ; 20(23): e2310556, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38386291

ABSTRACT

Skin injury repair is a dynamic process involving a series of interactions over time and space. Linking human physiological processes with materials' changes poses a significant challenge. To match the wound healing process, a spatiotemporal controllable biomimetic skin is developed, which comprises a three-dimensional (3D) printed membrane as the epidermis, a cell-containing hydrogel as the dermis, and a cytokine-laden hydrogel as the hypodermis. In the initial stage of the biomimetic skin repair wound, the membrane frame aids wound closure through pre-tension, while cells proliferate within the hydrogel. Next, as the frame disintegrates over time, cells released from the hydrogel migrate along the residual membrane. Throughout the process, continuous cytokines release from the hypodermis hydrogel ensures comprehensive nourishment. The findings reveal that in the rat full-thickness skin defect model, the biomimetic skin demonstrated a wound closure rate eight times higher than the blank group, and double the collagen content, particularly in the early repair process. Consequently, it is reasonable to infer that this biomimetic skin holds promising potential to accelerate wound closure and repair. This biomimetic skin with mechanobiological effects and spatiotemporal regulation emerges as a promising option for tissue regeneration engineering.


Subject(s)
Skin , Wound Healing , Animals , Rats , Hydrogels/chemistry , Biomimetics/methods , Biomimetic Materials/chemistry , Tissue Engineering/methods , Humans , Skin, Artificial , Rats, Sprague-Dawley , Printing, Three-Dimensional
18.
Small ; 20(31): e2310913, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38726952

ABSTRACT

Naturally occurring protein nanocages like ferritin are self-assembled from multiple subunits. Because of their unique cage-like structure and biocompatibility, there is a growing interest in their biomedical use. A multipurpose and straightforward engineering approach does not exist for using nanocages to make drug-delivery systems by encapsulating hydrophilic or hydrophobic drugs and developing vaccines by surface functionalization with a protein like an antigen. Here, a versatile engineering approach is described by mimicking the HIV-1 Gap polyprotein precursor. Various PREcursors of nanoCages (PREC) are designed and created by linking two ferritin subunits via a flexible linker peptide containing a protease cleavage site. These precursors can have additional proteins at their N-terminus, and their protease cleavage generates ferritin-like nanocages named protease-induced nanocages (PINCs). It is demonstrated that PINC formation allows concurrent surface decoration with a protein and hydrophilic or hydrophobic drug encapsulation up to fourfold more than the amount achieved using other methods. The PINCs/Drug complex is stable and efficiently kills cancer cells. This work provides insight into the precursors' design rules and the mechanism of PINCs formation. The engineering approach and mechanistic insight described here will facilitate nanocages' applications in drug delivery or as a platform for making multifunctional therapeutics like mosaic vaccines.


Subject(s)
Ferritins , Humans , Ferritins/chemistry , Surface Properties , HIV-1 , Hydrophobic and Hydrophilic Interactions , Drug Delivery Systems/methods , Nanostructures/chemistry , Biomimetic Materials/chemistry , Biomimetics/methods
19.
Small ; 20(35): e2400142, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38676334

ABSTRACT

Complex temporal molecular signals play a pivotal role in the intricate biological pathways of living organisms, and cells exhibit the ability to transmit and receive information by intricately managing the temporal dynamics of their signaling molecules. Although biomimetic molecular networks are successfully engineered outside of cells, the capacity to precisely manipulate temporal behaviors remains limited. In this study, the catalysis activity of isothermal DNA polymerase (DNAP) through combined use of molecular dynamics simulation analysis and fluorescence assays is first characterized. DNAP-driven delay in signal strand release ranged from 100 to 102 min, which is achieved through new strategies including the introduction of primer overhangs, utilization of inhibitory reagents, and alteration of DNA template lengths. The results provide a deeper insight into the underlying mechanisms of temporal control DNAP-mediated primer extension and DNA strand displacement reactions. Then, the regulated DNAP catalysis reactions are applied in temporal modulation of downstream DNA-involved reactions, the establishment of dynamic molecular signals, and the generation of barcodes for multiplexed detection of target genes. The utility of DNAP-based signal delay as a dynamic DNA nanotechnology extends beyond theoretical concepts and achieves practical applications in the fields of cell-free synthetic biology and bionic sensing.


Subject(s)
Biomimetics , DNA-Directed DNA Polymerase , DNA , DNA/chemistry , DNA/metabolism , DNA-Directed DNA Polymerase/metabolism , Biomimetics/methods , Molecular Dynamics Simulation , Biosensing Techniques/methods , Nanotechnology/methods
20.
Small ; 20(29): e2400666, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38368259

ABSTRACT

Compared to conventional radiotherapy (RT), FLASH-RT delivers ultra-high dose radiation, significantly reducing damage to normal tissue while guaranteeing the effect of cancer treatment. However, cancer recurrence and metastasis frequently occur after all RT due to the existence of intractable cancer stem cells (CSCs). To address this, a biomimetic nanoplatform (named TAFL) of tumor-derived exosome fusion liposomes is designed by co-loading aggregation-induced emission photothermal agents, TPE-BBT, and anti-cancer drugs, aspirin, aiming to clear CSCs for inhibiting cancer recurrence and metastasis after FLASH-RT therapy . Aspirin released in TAFL system triggered by laser irradiation can induce apoptosis and DNA damage of 4T1 CSCs, comprehensively downregulate their stemness phenotype, and inhibit their sphericity. Furthermore, the TPE-BBT mediated mild-photothermal therapy can alleviate the hypoxic tumor microenvironment, inhibit the DNA repair of CSCs, which further amplifies the effect of aspirin against CSCs, therefore reduces the effective dose of aspirin, making TAFL more biologically safe. In vivo experimental results demonstrated that decreased CSCs population mediated by TAFL system treatment significantly inhibited tumor recurrence and metastasis after FLASH-RT therapy. In summary, this TAFL system   provides a new idea for the future clinical application of FLASH-RT therapy.


Subject(s)
Aspirin , Breast Neoplasms , Neoplasm Metastasis , Neoplastic Stem Cells , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Animals , Female , Aspirin/pharmacology , Aspirin/therapeutic use , Cell Line, Tumor , Neoplasm Recurrence, Local , Mice , Humans , DNA Damage , Tumor Microenvironment/drug effects , Liposomes/chemistry , Apoptosis/drug effects , Biomimetics/methods , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Exosomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL