Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.737
Filtrer
1.
Biomaterials ; 313: 122804, 2025 Feb.
Article de Anglais | MEDLINE | ID: mdl-39236631

RÉSUMÉ

Insulin resistance and pancreatic ß-cell dysfunction are the main pathogenesis of type 2 diabetes mellitus (T2DM). However, insulin therapy and diabetes medications do not effectively solve the two problems simultaneously. In this study, a biomimetic oral hydrogen nanogenerator that leverages the benefits of edible plant-derived exosomes and hydrogen therapy was constructed to overcome this dilemma by modulating gut microbiota and ameliorating oxidative stress and inflammatory responses. Hollow mesoporous silica (HMS) nanoparticles encapsulating ammonia borane (A) were used to overcome the inefficiency of H2 delivery in traditional hydrogen therapy, and exosomes originating from ginger (GE) were employed to enhance biocompatibility and regulate intestinal flora. Our study showed that HMS/A@GE not only considerably ameliorated insulin resistance and liver steatosis, but inhibited the dedifferentiation of islet ß-cell and enhanced pancreatic ß-cell proportion in T2DM model mice. In addition to its antioxidant and anti-inflammatory effects, HMS/A@GE augmented the abundance of Lactobacilli spp. and tryptophan metabolites, such as indole and indole acetic acid, which further activated the AhR/IL-22 pathway to improve intestinal-barrier function and metabolic impairments. This study offers a potentially viable strategy for addressing the current limitations of diabetes treatment by integrating gut-microbiota remodelling with antioxidant therapies.


Sujet(s)
Antioxydants , Diabète de type 2 , Microbiome gastro-intestinal , Insulinorésistance , Cellules à insuline , Nanoparticules , Cellules à insuline/effets des médicaments et des substances chimiques , Cellules à insuline/métabolisme , Animaux , Diabète de type 2/métabolisme , Diabète de type 2/traitement médicamenteux , Antioxydants/pharmacologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Nanoparticules/composition chimique , Souris , Mâle , Matériaux biomimétiques/composition chimique , Matériaux biomimétiques/pharmacologie , Souris de lignée C57BL , Zingiber officinale/composition chimique , Silice/composition chimique , Exosomes/métabolisme , Biomimétique/méthodes , Stress oxydatif/effets des médicaments et des substances chimiques
2.
Biomaterials ; 313: 122801, 2025 Feb.
Article de Anglais | MEDLINE | ID: mdl-39236630

RÉSUMÉ

Chemoimmunotherapy is an emerging paradigm in the clinic for treating several malignant diseases, such as non-small cell lung cancer, breast cancer, and large B-cell lymphoma. However, the efficacy of this strategy is still restricted by serious adverse events and a high therapeutic termination rate, presumably due to the lack of tumor-targeted distribution of both chemotherapeutic and immunotherapeutic agents. Targeted drug delivery has the potential to address this issue. Among the most promising nanocarriers in clinical translation, liposomes have drawn great attention in cancer chemoimmunotherapy in recent years. Liposomes-enabled cancer chemoimmunotherapy has made significant progress in clinics, with impressive therapeutic outcomes. This review summarizes the latest preclinical and clinical progress in liposome-enabled cancer chemoimmunotherapy and discusses the challenges and future directions of this field.


Sujet(s)
Immunothérapie , Liposomes , Tumeurs , Liposomes/composition chimique , Humains , Immunothérapie/méthodes , Animaux , Tumeurs/thérapie , Tumeurs/traitement médicamenteux , Systèmes de délivrance de médicaments/méthodes , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/administration et posologie
3.
Biomaterials ; 313: 122775, 2025 Feb.
Article de Anglais | MEDLINE | ID: mdl-39241549

RÉSUMÉ

Acute Myocardial Infarction (AMI) has seen rising cases, particularly in younger people, leading to public health concerns. Standard treatments, like coronary artery recanalization, often don't fully repair the heart's microvasculature, risking heart failure. Advances show that Mesenchymal Stromal Cells (MSCs) transplantation improves cardiac function after AMI, but the harsh microenvironment post-AMI impacts cell survival and therapeutic results. MSCs aid heart repair via their membrane proteins and paracrine extracellular vesicles that carry microRNA-125b, which regulates multiple targets, preventing cardiomyocyte death, limiting fibroblast growth, and combating myocardial remodeling after AMI. This study introduces ultrasound-responsive phase-change bionic nanoparticles, leveraging MSCs' natural properties. These particles contain MSC membrane and microRNA-125b, with added macrophage membrane for stability. Using Ultrasound Targeted Microbubble Destruction (UTMD), this method targets the delivery of MSC membrane proteins and microRNA-125b to AMI's inflamed areas. This aims to enhance cardiac function recovery and provide precise, targeted AMI therapy.


Sujet(s)
Cellules souches mésenchymateuses , microARN , Infarctus du myocarde , Nanoparticules , Infarctus du myocarde/thérapie , Animaux , Nanoparticules/composition chimique , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , microARN/métabolisme , microARN/génétique , Mâle , Récupération fonctionnelle , Transplantation de cellules souches mésenchymateuses/méthodes , Humains , Matériaux biomimétiques/composition chimique , Matériaux biomimétiques/pharmacologie , Souris , Microbulles , Ondes ultrasonores
4.
Biomaterials ; 312: 122707, 2025 Jan.
Article de Anglais | MEDLINE | ID: mdl-39121729

RÉSUMÉ

Polypyrimidine tract-binding protein 1 (PTBP1) regulates numerous alternative splicing events during tumor progression and neurogenesis. Previously, PTBP1 downregulation was reported to convert astrocytes into functional neurons; however, how PTBP1 regulates astrocytic physiology remains unclear. In this study, we revealed that PTBP1 modulated glutamate uptake via ATP1a2, a member of Na+/K+-ATPases, and glutamate transporters in astrocytes. Ptbp1 knockdown altered mitochondrial function and energy metabolism, which involved PTBP1 regulating mitochondrial redox homeostasis via the succinate dehydrogenase (SDH)/Nrf2 pathway. The malfunction of glutamate transporters following Ptbp1 knockdown resulted in enhanced excitatory synaptic transmission in the cortex. Notably, we developed a biomimetic cationic triblock polypeptide system, i.e., polyethylene glycol44-polylysine30-polyleucine10 (PEG44-PLL30-PLLeu10) with astrocytic membrane coating to deliver Ptbp1 siRNA in vitro and in vivo, which approach allowed Ptbp1 siRNA to efficiently cross the blood-brain barrier and target astrocytes in the brain. Collectively, our findings suggest a framework whereby PTBP1 serves as a modulator in glutamate transport machinery, and indicate that biomimetic methodology is a promising route for in vivo siRNA delivery.


Sujet(s)
Astrocytes , Acide glutamique , Ribonucléoprotéines nucléaires hétérogènes , Homéostasie , Facteur-2 apparenté à NF-E2 , Protéine PTB , Petit ARN interférent , Animaux , Astrocytes/métabolisme , Acide glutamique/métabolisme , Protéine PTB/métabolisme , Protéine PTB/génétique , Facteur-2 apparenté à NF-E2/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , Souris , Transduction du signal , Membrane cellulaire/métabolisme , Souris de lignée C57BL , Mâle , Humains , Mitochondries/métabolisme
5.
Biomaterials ; 313: 122796, 2025 Feb.
Article de Anglais | MEDLINE | ID: mdl-39226654

RÉSUMÉ

Chemotherapy-induced cellular senescence leads to an increased proportion of cancer stem cells (CSCs) in breast cancer (BC), contributing to recurrence and metastasis, while effective means to clear them are currently lacking. Herein, we aim to develop new approaches for selectively killing senescent-escape CSCs. High CD276 (95.60%) expression in multidrug-resistant BC cells, facilitates immune evasion by low-immunogenic senescent escape CSCs. CALD1, upregulated in ADR-resistant BC, promoting senescent-escape of CSCs with an anti-apoptosis state and upregulating CD276, PD-L1 to promote chemoresistance and immune escape. We have developed a controlled-released thermosensitive hydrogel containing pH- responsive anti-CD276 scFV engineered biomimetic nanovesicles to overcome BC in primary, recurrent, metastatic and abscopal humanized mice models. Nanovesicles coated anti-CD276 scFV selectively fuses with cell membrane of senescent-escape CSCs, then sequentially delivers siCALD1 and ADR due to pH-responsive MnP shell. siCALD1 together with ADR effectively induce apoptosis of CSCs, decrease expression of CD276 and PD-L1, and upregulate MHC I combined with Mn2+ to overcome chemoresistance and promote CD8+T cells infiltration. This combined therapeutic approach reveals insights into immune surveillance evasion by senescent-escape CSCs, offering a promising strategy to immunotherapy effectiveness in cancer therapy.


Sujet(s)
Tumeurs du sein , Vieillissement de la cellule , Résistance aux médicaments antinéoplasiques , Cellules souches tumorales , Humains , Animaux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/thérapie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Femelle , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Souris , Matériaux biomimétiques/composition chimique , Matériaux biomimétiques/pharmacologie , Génie génétique/méthodes , Doxorubicine/pharmacologie , Doxorubicine/usage thérapeutique , Nanoparticules/composition chimique , Anticorps à chaîne unique/composition chimique , Échappement de la tumeur à la surveillance immunitaire/effets des médicaments et des substances chimiques , Antigène CD274/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Biomimétique/méthodes , Antigènes B7
6.
Small Methods ; : e2400968, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39351807

RÉSUMÉ

Solid-state electrolytes (SSEs) based on metal-organic frameworks (MOFs) are an ideal material for constructing high-performance lithium metal batteries (LMBs). However, the low ion conductivity and poor interface contact (especially at low temperatures) still seriously hinder its further application. Herein, inspired by the Na+/K+ conduction in biology systems, a series (NH2, OH, NH-(CH2)3-SO3H)-modified MIL-53-X as SSEs is reported. These functional groups are similar to anions suspended in biological ion channels, partially repelling anions while allowing cations to be effectively transported through pore channels. Subsequently, MIL-53-X with hierarchical pore structure (H-MIL-53-X) is obtained by introducing lauric acid as a regulator, and then the effects of structural design and morphology control on its performance are explored. The conductivity of H-MIL-53-NH-SO3Li with multi-level pore structure and modified by sulfonic acid groups reached 2.2 × 10-3 S cm-1 at 25 °C, lithium-ion transference number of 0.78. Besides, the H-MIL-53-NH-SO3Li still has an excellent conductivity of 10-4 S cm-1 at -40 °C. Additionally, LiFePO4/Li batteries equipped with H-MIL-53-NH-SO3Li SSEs could operate stably for over 200 cycles at 0.1 C. The strategy of combining structural and morphological design of MOFs with biomimetic ion channels opens new avenues for the design of high-performance SSEs.

7.
Adv Funct Mater ; 34(35): 2305040, 2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-39355086

RÉSUMÉ

Artificial spider silk is an attractive material for many technical applications since it is a biobased fiber that can be produced under ambient conditions but still outcompetes synthetic fibers (e.g., Kevlar) in terms of toughness. Industrial use of this material requires bulk-scale production of recombinant spider silk proteins in heterologous host and replication of the pristine fiber's mechanical properties. High molecular weight spider silk proteins can be spun into fibers with impressive mechanical properties, but the production levels are too low to allow commercialization of the material. Small spider silk proteins, on the other hand, can be produced at yields that are compatible with industrial use, but the mechanical properties of such fibers need to be improved. Here, the literature on wet-spinning of artificial spider silk fibers is summarized and analyzed with a focus on mechanical performance. Furthermore, several strategies for how to improve the properties of such fibers, including optimized protein composition, smarter spinning setups, innovative protein engineering, chemical and physical crosslinking as well as the incorporation of nanomaterials in composite fibers, are outlined and discussed.

8.
Biomaterials ; 314: 122821, 2024 Sep 08.
Article de Anglais | MEDLINE | ID: mdl-39357151

RÉSUMÉ

The treatment of osteochondral joint lesions requires the regeneration of both articular cartilage and subchondral bone tissue. Scaffold-based strategies aimed at mimicking the native osteochondral structure have been explored with mixed results. The aim of this study was to evaluate the regenerative potential of a tri-layered osteochondral cell-free scaffold in a large animal model at both 6 and 12 months of follow-up. Bilateral critical-sized osteochondral defects were created in 22 sheep. One defect was filled with the scaffold, whereas the contralateral was left empty. The repair tissue quality was evaluated at 6 and 12 months of follow-up in terms of macroscopic appearance, histology, trabecular bone formation, and inflammation grade. The mean global ICRS II score in the scaffold and control groups was 41 ± 11 vs 30 ± 6 at 6 months (p = 0.004) and 54 ± 13 vs 37 ± 11 at 12 months (p = 0.002), respectively. A higher percentage of bone was found in the treatment group compared to controls both at 6 (BV/TV 48.8 ± 8.6 % vs 37.4 ± 9.5 %, respectively; p < 0.001) and 12 months (BV/TV 51.8 ± 8.8 % vs 42.1 ± 12.6 %, respectively; p = 0.023). No significant levels of inflammation were seen. These results demonstrated the scaffold safety and potential to regenerate both cartilage and subchondral tissues in a large animal model of knee osteochondral lesions.

9.
Adv Mater ; : e2408361, 2024 Oct 02.
Article de Anglais | MEDLINE | ID: mdl-39358930

RÉSUMÉ

Arsenic agents have shown great potential in fighting leukemia, but are poorly known in treating solid tumors, mainly ascribing to the rapid clearance and low targeting ability. It is reported that morphology modulation can enhance the interaction between nanoparticles and cell membrane. Herein, a dismountable protein corona-modified virus-like manganese-arsenic nanomedicine (vMnAs@HR) is rationally proposed for realizing safe and targeted delivery and synergistic arsenotherapy. The virus-like manganese-arsenic nanoparticle (vMnAs) is constructed followed by modification of a temporary R848-loaded HDL (HR) protein corona. Upon intravenous injection, the HR protein corona is stable and actively targeted to tumor tissue by taking advantage of the interaction between HDL and its receptor SR-BI. Intriguingly, upon accumulated in the tumor, HR can be jettisoned and interacted with macrophages for proinflammatory phenotype modulation. The re-exposed vMnAs can efficiently enhance endocytosis by taking advantage of the rationally designed spiky morphology. Moreover, the released double-stranded DNA (dsDNA) and manganese ions during tumor cell apoptosis can cooperatively activate cyclic guanosine monophosphate adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway of DCs for systematic immune activation. It is anticipated that this morphology-transformable nanomedicine can realize safe and efficient arsenic delivery for synergistic arsenotherapy.

10.
ACS Nano ; 2024 Oct 03.
Article de Anglais | MEDLINE | ID: mdl-39360741

RÉSUMÉ

A cell membrane-derived vesicle (MV) that has cell-mimicking features with characteristic functionalities holds vast appeal for biomimetic nanomedicine and drug delivery but suffers from a major limitation of innate fragility and poor stability. Herein, we report a lipid-anchoring strategy for stabilizing MV for enhanced drug delivery. An array of amphiphilic mono-acyl phosphatidylcholines (MPCs) with specific hydrophobic moieties are synthesized and readily engineered on MV based on their commendable aqueous solubility and efficient membrane insertability. Incorporation of MPCs containing rigid ring structures in the hydrophobic segment demonstrates the potency of stabilizing MV by the combined ordering and condensing effects. The optimized MPC-stabilized MV exhibits prolonged circulation in the bloodstream, elevated accumulation within a tumor, and enhanced therapeutic effects of chemotherapeutic and photothermal drugs. Moreover, doxorubicin-loaded MV, engineered with mono-all-trans retinoyl phosphatidylcholine as an MV stabilizer and a therapeutic prodrug, potently suppresses growth and metastasis of high-stemness tumors.

11.
Chembiochem ; : e202400674, 2024 Oct 02.
Article de Anglais | MEDLINE | ID: mdl-39356249

RÉSUMÉ

Disulfide-bonded peptides and proteins, including hormones, toxins, growth factors, and others, are abundant in living organisms. These molecules play crucial physiological roles such as regulating cell and organism growth, development, and metabolism. They have also found widespread applications as drugs or tool molecules in biomedical and pharmaceutical research. However, the chemical synthesis of disulfide-bonded proteins is complicated by the challenges associated with their folding. This review focuses on the latest advancements in disulfide-bonded peptide and protein folding technologies. Particularly, it highlights biomimetic folding strategies that emulate the naturally occurring oxidative folding processes in nature. These strategies include chaperone-assisted folding, glycosylation-assisted folding, and organic-based oxidative folding methods. The review also anticipates future directions in folding technology. Such research offers innovative approaches for the chemical synthesis of complex proteins that are otherwise difficult-to-fold.

12.
J Control Release ; 376: 67-78, 2024 Oct 09.
Article de Anglais | MEDLINE | ID: mdl-39368706

RÉSUMÉ

Glioblastoma multiforme (GBM), the most aggressive intracranial neoplasm, remains incurable at present, primarily due to drug resistance, which significantly contributes to elevated recurrence rates and dismal prognosis. Signal transducer and activator of transcription 3 (STAT3) is a critical gene closely associated with GBM drug resistance and the progression of GBM stem cells (GSCs), making it a promising therapeutic target. In this study, we developed cancer cell membrane-cloaked biomimetic nanoparticles to deliver STAT3 siRNA to reverse drug resistance in homologous GBM. These biomimetic nanoparticles leverage homotypic targeting, rapid endosome escape, and fast siRNA release, leading to efficient in vitro STAT3 knockdown in both temozolomide-resistant U251-TR cells and X01 GSCs. Moreover, benefited from the membrane functionalization, significant prolonged blood circulation, improved blood brain barrier (BBB) penetration and GBM tumor accumulation are achieved by these siRNA biomimetic nanoparticles. Importantly, these nanoparticles effectively inhibit tumor proliferation, significantly extending median survival time in orthotopic U251-TR (43.5 d versus 20 d for PBS control) and X01 GSC-bearing mouse xenografts (52 d versus 19.5 d for PBS control). Altogether, this biomimetic siRNA platform offers a promising strategy for gene therapy targeting drug-resistant GBM.

13.
Colloids Surf B Biointerfaces ; 245: 114290, 2024 Oct 02.
Article de Anglais | MEDLINE | ID: mdl-39383582

RÉSUMÉ

Proteoglycans (PG) is crucial for regulating collagen formation and mineralization during bone tissue development. A wide variety of PG-modified collagen scaffolds have been proposed for bone engineering application to promote biological responses and work as artificial matrices that guide tissue regeneration. However, poor performance of theses biomaterials against infections has led to an unmet need for clinical prevention. Therefore, we utilized proanthocyanidins (PA) to simulate the functions of PG, including mediating the collagen assembly and intrafibrillar mineralization, to optimize scaffolds performance. The excellent antibacterial properties of PA can endow the scaffolds with anti-infection effects in the process of tissue regeneration. When PA was added during fibrillogenesis, the collagen fibrils appeared irregular aggregation and the mineralization degree was reduced. In contrast, the addition of PA after collagen self-assembly improved the latter's ability to act as a deposition template and remarkably promoted mineral ions infiltration, thus enhancing intrafibrillar mineralization. The PA-modified scaffold displayed a highly hydrophilicity behaviour and long-term resistance to degradation. The sustained release of PA effectively inhibited the activity of Staphylococcus aureus. The scaffold also showed excellent biocompatibility and improved bone regeneration in calvarial critical-size defect models. The application of PA enables a dual-function scaffold with favourable intrafibrillar mineralization and anti-bacterial properties for bone regeneration.

14.
Eur J Pharm Biopharm ; : 114527, 2024 Oct 07.
Article de Anglais | MEDLINE | ID: mdl-39383975

RÉSUMÉ

The efficient and secure delivery of intravenous chemotherapeutic agents across the blood-brain barrier (BBB) to the precise location of a brain tumor is a crucial element in glioma treatment. Herein, we introduce a biomimetic nanoplatform (T7-M-C/S) comprising a core made up of irinotecan hydrochloride (CPT11) and its bioactive metabolite, 7-Ethyl-10-hydroxycamptothecin (SN38), surrounded by a layer of T7-peptide-modified macrophage membrane. CPT11 spontaneously assembles with SN38 into stable and water-dispersible nanoparticles (C/S), greatly enhancing the water solubility of SN38. The integration of the modified peptide with the inherent proteins expressed by macrophage cells confers the nanoplatform with enhanced bioavailability and robust glioma-targeting abilities, ultimately resulting in superior therapeutic outcomes. These discoveries highlight a drug delivery system characterized by a high drug loading capacity, leveraging the macrophage membrane, and promising significant potential for glioma treatment.

15.
ACS Nano ; 2024 Oct 05.
Article de Anglais | MEDLINE | ID: mdl-39367850

RÉSUMÉ

Sepsis-associated encephalopathy (SAE) is a devastating complication of sepsis, affecting approximately 70% of patients with sepsis in intensive care units (ICU). Although the pathophysiological mechanisms remain elusive, sepsis is typically accompanied by systemic inflammatory response syndrome (SIRS) and hyper-oxidative conditions. Here, we introduce a biomimetic nanomodulator (mAOI NP) that specifically targets inflammation site and simultaneously regulates oxidative and inflammatory stresses. mAOI NPs are constructed using metal-coordinated polyphenolic antioxidants (tannic acid) and flavonoid quercetin, which are then coated with macrophage membrane to enhance pharmacokinetics and enable SAE targeting. In a cecal ligation and puncture (CLP)-induced severe sepsis model, mAOI NPs effectively mitigate oxidative stress by purging reactive oxygen species, repairing mitochondrial damage and activating the Nrf2/HO-1 signaling pathway; while polarizing M1 macrophages or microglia toward anti-inflammatory M2 subtype. mAOI NPs potently inhibit sepsis progress, prolong overall survival from 25 to 66% and enhance learning and memory capabilities in SAE mice. Further proteomics analysis reveals that mAOI NPs modulate neurodevelopment processes related to learning and memory formation while also exerting anti-inflammatory and antioxidative effects on brain tissue responses associated with SAE pathology. This study offers significant potential for improving patient outcomes and revolutionizing the treatment landscape for this devastating complication of sepsis.

16.
Macromol Biosci ; : e2400146, 2024 Oct 07.
Article de Anglais | MEDLINE | ID: mdl-39374341

RÉSUMÉ

A major limitation in the development of mucosal drug delivery systems is the design of in vitro models that accurately reflect in vivo conditions. Traditionally, models seek to mimic characteristics of physiological mucus, often focusing on property-specific trial metrics such as rheological behavior or diffusion of a nanoparticle of interest. Despite the success of these models, translation from in vitro results to in vivo trials is limited. As a result, several authors have called for work to develop standardized testing methodologies and characterize the influence of model properties on drug delivery performance. To this end, a series of trials is performed on 12 mucomimetic hydrogels reproduced from literature. Experiments show that there is no consistent correlation between barrier performance and rheological or microstructural properties of the tested mucomimetic hydrogels. In addition, the permeability of both mucopenetrating and mucoadhesive nanoparticles is assessed, revealing non-obvious variations in barrier properties such as the relative contributions of electrostatic and hydrophobic interactions in different models. These results demonstrate the limitations of predicting mucomimetic behavior with common characterization techniques and highlight the importance of testing barrier performance with multiple nanoparticle formulations.

17.
J Biomed Mater Res A ; 2024 Oct 04.
Article de Anglais | MEDLINE | ID: mdl-39367651

RÉSUMÉ

Enhancing the biocompatibility and mechanical stability of small diameter vascular scaffolds remain significant challenges. To address this challenge, small-diameter tubular structures were electrospun from silk fibroin (SF) from silk textile industry discarded materials to generate bilayer scaffolds that mimic native blood vessels, but derived from a sustainable natural material resource. The inner layer was obtained by directly dissolving SF in formic acid, while the middle layer (SF-M) was achieved through aqueous concentration of the protein. Structural and biological properties of each layer as well as the bilayer were evaluated. The inner layer exhibited nano-scale fiber diameters and 57.9% crystallinity, and degradation rates comparable with the SF-M layer. The middle layer displayed micrometer-scale fibers diameters with an ultimate extension of about 274%. Both layers presented contact angles suitable for cell growth and cytocompatibility, while the bilayer material displayed an intermediate mechanical response and a reduced enzymatic degradation rate when compared to each individual layer. The bilayer material emulates many of the characteristics of native small-diameter vessels, thereby suggesting further studies towards in vivo opportunities.

18.
J Nanobiotechnology ; 22(1): 604, 2024 Oct 06.
Article de Anglais | MEDLINE | ID: mdl-39370518

RÉSUMÉ

Inspired by the concept of "natural camouflage," biomimetic drug delivery systems have emerged to address the limitations of traditional synthetic nanocarriers, such as poor targeting, susceptibility to identification and clearance, inadequate biocompatibility, low permeability, and systemic toxicity. Biomimetic nanocarriers retain the proteins, nucleic acids, and other components of the parent cells. They not only facilitate drug delivery but also serve as communication media to inhibit tumor cells. This paper delves into the communication mechanisms between various cell-derived biomimetic nanocarriers, tumor cells, and the tumor microenvironment, as well as their applications in drug delivery. In addition, the additional communication capabilities conferred on the modified biomimetic nanocarriers, such as targeting and environmental responsiveness, are outlined. Finally, we propose future development directions for biomimetic nanocarriers, hoping to inspire researchers in their design efforts and ultimately achieve clinical translation.


Sujet(s)
Matériaux biomimétiques , Vecteurs de médicaments , Nanoparticules , Tumeurs , Microenvironnement tumoral , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Humains , Tumeurs/traitement médicamenteux , Matériaux biomimétiques/composition chimique , Matériaux biomimétiques/pharmacologie , Vecteurs de médicaments/composition chimique , Nanoparticules/composition chimique , Animaux , Systèmes de délivrance de médicaments/méthodes , Communication cellulaire/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Biomimétique/méthodes
19.
Article de Anglais | MEDLINE | ID: mdl-39375966

RÉSUMÉ

Clinically relevant matrices such as human blood and serum can cause substantial interference in biosensing measurements, severely compromising the effectiveness of the sensors. We report the characterization of a positively charged lipid membrane that has demonstrated unique features to suppress the nonspecific signal for antifouling effects by using SPR, fluorescence recovery after photobleaching (FRAP), and MALDI-TOF-MS. The ethylphosphocholine (EPC) lipid membrane proved to be exceptionally effective at reducing irreversible interactions from human serum on a Protein A surface. The membrane formation conditions and their effects on membrane fluidity and mobility were characterized for understanding the antifouling functions when various capture molecules were immobilized. Specifically, EPC lipid membranes on a Protein A substrate appear to exhibit a strong interaction, likely through the electrostatic effect with the negatively charged proteins that resulted in a stable hydration layer. The strong interaction also limited lipid mobility, contributing to a robust, protective interface that remained undamaged in undiluted serum. Tailoring a surface with antifouling lipid membranes allows for a range of biosensing applications in highly complex biological media.

20.
ACS Nano ; 2024 Oct 10.
Article de Anglais | MEDLINE | ID: mdl-39388302

RÉSUMÉ

Integrating self-healing capabilities into epidermal electrodes is crucial to improving their reliability and longevity. Self-healing nanofibrous materials are considered an ideal candidate for constructing ultrathin, long-lasting wearable epidermal electrodes due to their lightweight and high breathability. However, due to the strong interaction between fibers, self-healing nanofiber membranes cannot exist stably. Therefore, the development of self-healing and breathable nanofibrous epidermal electrodes still remains a major challenge. Here, a hierarchical confinement strategy that combines molecular and spatial confinement to overcome supramolecular hydrogen bonding between self-healing nanofibers is reported, and an ultrathin self-healing nanofibrous epidermal electrode with a neural net-like structure is developed. It can achieve real-time monitoring of electrophysiological signals through long-term conformal attachment to skin or plants and has no adverse effects on skin health or plant growth. Given the almost imperceptible nature of epidermal electrodes to users and plants, it lays the foundation for the development of biocompatible, self-healing, wearable, flexible electronics.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE