Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
J Nanobiotechnology ; 19(1): 385, 2021 Nov 22.
Article in English | MEDLINE | ID: mdl-34809623

ABSTRACT

Demineralization of hard tooth tissues leads to dental caries, which cause health problems and economic burdens throughout the world. A biomimetic mineralization strategy is expected to reverse early dental caries. Commercially available anti-carious mineralizing products lead to inconclusive clinical results because they cannot continuously replenish the required calcium and phosphate resources. Herein, we prepared a mineralizing film consisting of hydroxypropylmethylcellulose (HPMC) and polyaspartic acid-stabilized amorphous calcium phosphate (PAsp-ACP) nanoparticles. HPMC which contains multiple hydroxyl groups is a film-forming material that can be desiccated to form a dry film. In a moist environment, this film gradually changes into a gel. HPMC was used as the carrier of PAsp-ACP nanoparticles to deliver biomimetic mineralization. Our results indicated that the hydroxyl and methoxyl groups of HPMC could assist the stability of PAsp-ACP nanoparticles and maintain their biomimetic mineralization activity. The results further demonstrated that the bioinspired mineralizing film induced the early mineralization of demineralized dentin after 24 h with increasing mineralization of the whole demineralized dentin (3-4 µm) after 72-96 h. Furthermore, these results were achieved without any cytotoxicity or mucosa irritation. Therefore, this mineralizing film shows promise for use in preventive dentistry due to its efficient mineralization capability.


Subject(s)
Biomimetic Materials , Calcium Phosphates , Dental Caries/metabolism , Hypromellose Derivatives , Tooth Calcification/drug effects , Animals , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacokinetics , Biomimetic Materials/pharmacology , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacokinetics , Calcium Phosphates/pharmacology , Cells, Cultured , Dentin/drug effects , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Hypromellose Derivatives/chemistry , Hypromellose Derivatives/pharmacology , Male , Mice , Nanoparticle Drug Delivery System , Nanoparticles , Rabbits
2.
J Nanobiotechnology ; 19(1): 374, 2021 Nov 17.
Article in English | MEDLINE | ID: mdl-34789284

ABSTRACT

BACKGROUND: Intimal hyperplasia caused by vascular injury is an important pathological process of many vascular diseases, especially occlusive vascular disease. In recent years, Nano-drug delivery system has attracted a wide attention as a novel treatment strategy, but there are still some challenges such as high clearance rate and insufficient targeting. RESULTS: In this study, we report a biomimetic ROS-responsive MM@PCM/RAP nanoparticle coated with macrophage membrane. The macrophage membrane with the innate "homing" capacity can superiorly regulate the recruitment of MM@PCM/RAP to inflammatory lesion to enhance target efficacy, and can also disguise MM@PCM/RAP nanoparticle as the autologous cell to avoid clearance by the immune system. In addition, MM@PCM/RAP can effectively improve the solubility of rapamycin and respond to the high concentration level of ROS accumulated in pathological lesion for controlling local cargo release, thereby increasing drug availability and reducing toxic side effects. CONCLUSIONS: Our findings validate that the rational design, biomimetic nanoparticles MM@PCM/RAP, can effectively inhibit the pathological process of intimal injury with excellent biocompatibility.


Subject(s)
Hyperplasia/metabolism , Macrophages/cytology , Nanoparticle Drug Delivery System , Reactive Oxygen Species/metabolism , Tunica Intima , Animals , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacokinetics , Biomimetic Materials/pharmacology , Cell Membrane/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Male , Mice , Mice, Inbred C57BL , Nanoparticle Drug Delivery System/chemistry , Nanoparticle Drug Delivery System/metabolism , Sirolimus/chemistry , Sirolimus/pharmacokinetics , Sirolimus/pharmacology , Tunica Intima/drug effects , Tunica Intima/metabolism , Zebrafish
3.
J Am Chem Soc ; 143(43): 18029-18040, 2021 11 03.
Article in English | MEDLINE | ID: mdl-34664942

ABSTRACT

The chemical topology is a unique dimension for protein engineering, yet the topological diversity and architectural complexity of proteins remain largely untapped. Herein, we report the biosynthesis of complex topological proteins using a rationally engineered, cross-entwining peptide heterodimer motif derived from p53dim (an entangled homodimeric mutant of the tetramerization domain of the tumor suppressor protein p53). The incorporation of an electrostatic interaction at specific sites converts the p53dim homodimer motif into a pair of heterodimer motifs with high specificity for directing chain entanglement upon folding. Its combination with split-intein-mediated ligation and/or SpyTag/SpyCatcher chemistry facilitates the programmed synthesis of protein heterocatenane or [n]catenanes in cells, leading to a general and modular approach to complex protein catenanes containing various proteins of interest. Concatenation enhances not only the target protein's affinity but also the in vivo stability as shown by its prolonged circulation time in blood. As a proof of concept, artificial antibodies have been developed by embedding a human epidermal growth factor receptor 2-specific affibody onto the [n]catenane scaffolds and shown to exhibit a higher affinity and a better pharmacokinetic profile than the wild-type affibody. These results suggest that topology engineering holds great promise in the development of therapeutic proteins.


Subject(s)
Antibodies/chemistry , Biomimetic Materials/metabolism , Catenanes/metabolism , Peptide Fragments/metabolism , Recombinant Fusion Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Animals , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacokinetics , Catenanes/chemistry , Catenanes/pharmacokinetics , Cell Line, Tumor , Female , Humans , Mice, Inbred BALB C , Peptide Fragments/chemistry , Peptide Fragments/pharmacokinetics , Proof of Concept Study , Protein Domains , Protein Engineering , Protein Structure, Quaternary , Receptor, ErbB-2/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacokinetics , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/pharmacokinetics
4.
J Nanobiotechnology ; 19(1): 339, 2021 Oct 24.
Article in English | MEDLINE | ID: mdl-34689761

ABSTRACT

BACKGROUND: Biomimetic nanotechnology-based RNA interference (RNAi) has been successful in improving theranostic efficacy in malignant tumors. Its integration with hybrid biomimetic membranes made of natural cell membranes fused with liposomal membranes is mutually beneficial and extends their biofunctions. However, limited research has focused on engineering such biomimetics to endow them with unique properties and functions, in particular, those essential for a "smart" drug delivery system, such as a tumor microenvironment (TME)-activated multifunctional biomimetic nanoplatform. RESULTS: Herein, we utilized an integrated hybrid nanovesicle composed of cancer cell membranes (Cm) and matrix metallopeptidase 9 (MMP-9)-switchable peptide-based charge-reversal liposome membranes (Lipm) to coat lipoic acid-modified polypeptides (LC) co-loaded with phosphoglycerate mutase 1 (PGAM1) siRNA (siPGAM1) and DTX. The nanovesicle presented a negatively charged coating (citraconic anhydride-grafted poly-L-lysine, PC) in the middle layer for pH-triggered charge conversion functionalization. The established chemotherapeutic drug (DTX) co-delivery system CLip-PC@CO-LC nanoparticles (NPs) have a particle size of ~ 193 nm and present the same surface proteins as the Cm. Confocal microscopy and flow cytometry results indicated a greater uptake of MMP-9-treated CLip-PC@CO-LC NPs compared with that of the CLip-PC@CO-LC NPs without MMP-9 pretreatment. The exposure to MMP-9 activated positively charged cell-penetrating peptides on the surface of the hybrid nanovesicles. Moreover, pH triggered membrane disruption, and redox triggered DTX and siRNA release, leading to highly potent target-gene silencing in glycolysis and chemotherapy with enhanced antiproliferation ability. The biodistribution results demonstrated that the CLip-PC@LC-DiR NPs accumulated in the tumor owing to a combination of long blood retention time, homologous targeting ability, and TME-activated characteristics. The CLip-PC@CO-LC NPs led to more effective tumor growth inhibition than the DTX and free siPGAM1 formulations. CONCLUSIONS: TME-activated cancer cell membrane-liposome integrated hybrid NPs provide an encouraging nanoplatform that combines RNAi with chemotherapy for precise treatment of non-small cell lung cancer.


Subject(s)
Antineoplastic Agents , Biomimetic Materials , Cell Membrane/chemistry , Liposomes , Tumor Microenvironment/drug effects , A549 Cells , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacokinetics , Biomimetic Materials/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Drug Delivery Systems , Glycolysis , Humans , Liposomes/chemistry , Liposomes/pharmacokinetics , Lung Neoplasms/metabolism , Mice , Mice, Nude , Nanoparticles/chemistry , Nanoparticles/metabolism
5.
Drug Deliv ; 28(1): 1237-1255, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34142930

ABSTRACT

Nanotechnology provides synthetic carriers for cancer drug delivery that protect cargos from degradation, control drug release and increase local accumulation at tumors. However, these non-natural vehicles display poor tumor targeting and potential toxicity and are eliminated by the immune system. Recently, biomimetic nanocarriers have been widely developed based on the concept of 'mimicking nature.' Among them, cell-derived biomimetic vehicles have become the focus of bionics research because of their multiple natural functions, such as low immunogenicity, long circulation time and targeting ability. Cell membrane-coated carriers and extracellular vesicles are two widely used cell-based biomimetic materials. Here, this review summarizes the latest progress in the application of these two biomimetic carriers in targeted cancer therapy. Their properties and performance are compared, and their future challenges and development prospects are discussed.


Subject(s)
Biomimetic Materials/pharmacology , Biomimetics/methods , Cell Membrane/metabolism , Drug Carriers/pharmacology , Extracellular Vesicles/metabolism , Biomimetic Materials/pharmacokinetics , Chemistry, Pharmaceutical , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Humans , Nanoparticles/chemistry , Neoplasms/drug therapy
6.
Eur J Pharm Biopharm ; 158: 382-389, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33309845

ABSTRACT

Interaction of conventional drug delivery systems such as polymeric or lipid based nano- and microparticles with the in vivo milieu has garnered significant interest, primarily to orchestrate immune escape and/or improve targeting. Surface modification with targeting ligands has been heavily relied upon for the mentioned purpose in the recent years. However, the surface modified particles can also activate the immune system. Large-scale manufacturing can also be a challenge, as surface modification needs to be reproducible. Furthermore, in vivo, the targeting is dependent on the receptor expression density and number of target sites, which adds to the pharmacokinetic variability of the constructs. An evolving paradigm to overcome complications of surface functionalization is the incorporation of bio-inspired topographies into these conventional delivery systems to enable them to better interact with biological systems. Biomimetic delivery systems combine the unique surface composition of cells or cell membranes, and versatility of synthetic nanoparticles. In this review, we focus on one such delivery system, silica particles, and explore their interaction with different biological membranes.


Subject(s)
Antineoplastic Agents/administration & dosage , Biomimetic Materials/chemistry , Drug Carriers/chemistry , Neoplasms/drug therapy , Silicon Dioxide/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Biomimetic Materials/pharmacokinetics , Cell Membrane/metabolism , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Disease Models, Animal , Drug Carriers/pharmacokinetics , Drug Liberation , Humans , Nanoparticles/chemistry , Silicon Dioxide/pharmacokinetics , Surface Properties
7.
Theranostics ; 10(25): 11607-11621, 2020.
Article in English | MEDLINE | ID: mdl-33052236

ABSTRACT

The hypoxic microenvironment in solid tumors severely limits the efficacy of photodynamic therapy (PDT). Therefore, the development of nanocarriers co-loaded with photosensitizers and oxygen, together with imaging guidance ability, is of great significance in cancer therapy. However, previously reported synthetic methods for these multi-functional probes are complicated, and the raw materials used are toxic. Methods: Herein, the human endogenous protein, hemoglobin (Hb), was used for the simultaneous biomimetic synthesis of Gd-based nanostructures and co-loading of Chlorine e6 (Ce6) and oxygen for alleviating the hypoxic environment of tumors and accomplishing magnetic resonance imaging (MRI)-guided enhanced PDT. The Gd@HbCe6-PEG nanoprobes were synthesized via a green and protein biomimetic approach. The physicochemical properties, including relaxivity, oxygen-carrying/release capability, and PDT efficacy of Gd@HbCe6-PEG, were measured in vitro and in vivo on tumor-bearing mice after intravenous injection. Morphologic and functional MRI were carried out to evaluate the efficacy of PDT. Results: The results demonstrated the successful synthesis of compact Gd@HbCe6-PEG nanostructures with desired multi-functionalities. Following treatment with the nanoparticles, the embedded MR moiety was effective in lighting tumor lesions and guiding therapy. The oxygen-carrying capability of Hb after biomimetic synthesis was confirmed by spectroscopic analysis and oxygen detector in vitro. Further, tumor oxygenation for alleviating tumor hypoxia in vivo after intravenous injection of Gd@HbCe6-PEG was verified by photoacoustic imaging and immunofluorescence staining. The potent treatment efficacy of PDT on early-stage was observed by the morphologic and functional MR imaging. Importantly, rapid renal clearance of the particles was observed after treatment. Conclusion: In this study, by using a human endogenous protein, we demonstrated the biomimetic synthesis of multi-functional nanoprobes for simultaneous tumor oxygenation and imaging-guided enhanced PDT. The therapeutic efficacy could be quantitatively confirmed at 6 h post PDT with diffusion-weighted imaging (DWI).


Subject(s)
Antineoplastic Agents/administration & dosage , Metal Nanoparticles/administration & dosage , Neoplasms/drug therapy , Photochemotherapy/methods , Theranostic Nanomedicine/methods , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Biomimetic Materials/administration & dosage , Biomimetic Materials/chemical synthesis , Biomimetic Materials/pharmacokinetics , Cell Line, Tumor/transplantation , Chlorophyllides , Diffusion Magnetic Resonance Imaging , Disease Models, Animal , Female , Gadolinium/administration & dosage , Gadolinium/chemistry , Green Chemistry Technology , Hemoglobins/administration & dosage , Hemoglobins/chemistry , Humans , Injections, Intravenous , Metal Nanoparticles/chemistry , Mice , Molecular Probes/administration & dosage , Molecular Probes/chemical synthesis , Molecular Probes/pharmacokinetics , Neoplasms/diagnostic imaging , Oxygen/administration & dosage , Oxygen/chemistry , Photoacoustic Techniques , Porphyrins/administration & dosage , Porphyrins/chemistry , Tumor Hypoxia/drug effects , Tumor Microenvironment/drug effects
8.
J Nanobiotechnology ; 18(1): 115, 2020 Aug 20.
Article in English | MEDLINE | ID: mdl-32819405

ABSTRACT

BACKGROUND: Glucocorticoids (GCs) show powerful treatment effect on rheumatoid arthritis (RA). However, the clinical application is limited by their nonspecific distribution after systemic administration, serious adverse reactions during long-term administration. To achieve better treatment, reduce side effect, we here established a biomimetic exosome (Exo) encapsulating dexamethasone sodium phosphate (Dex) nanoparticle (Exo/Dex), whose surface was modified with folic acid (FA)-polyethylene glycol (PEG)-cholesterol (Chol) compound to attain FPC-Exo/Dex active targeting drug delivery system. RESULTS: The size of FPC-Exo/Dex was 128.43 ± 16.27 nm, with a polydispersity index (PDI) of 0.36 ± 0.05, and the Zeta potential was - 22.73 ± 0.91 mV. The encapsulation efficiency (EE) of the preparation was 10.26 ± 0.73%, with drug loading efficiency (DLE) of 18.81 ± 2.05%. In vitro study showed this system displayed enhanced endocytosis and excellent anti-inflammation effect against RAW264.7 cells by suppressing pro-inflammatory cytokines and increasing anti-inflammatory cytokine. Further biodistribution study showed the fluorescence intensity of FPC-Exo/Dex was stronger than other Dex formulations in joints, suggesting its enhanced accumulation to inflammation sites. In vivo biodistribution experiment displayed FPC-Exo/Dex could preserve the bone and cartilage of CIA mice better and significantly reduce inflamed joints. Next in vivo safety evaluation demonstrated this biomimetic drug delivery system had no obvious hepatotoxicity and exhibited desirable biocompatibility. CONCLUSION: The present study provides a promising strategy for using exosome as nanocarrier to enhance the therapeutic effect of GCs against RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , Biomimetic Materials , Dexamethasone , Exosomes , Nanoparticles , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacokinetics , Arthritis, Rheumatoid/pathology , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacokinetics , Cholesterol/analogs & derivatives , Cholesterol/chemistry , Dexamethasone/chemistry , Dexamethasone/pharmacokinetics , Exosomes/chemistry , Exosomes/metabolism , Folic Acid/chemistry , Joints/metabolism , Joints/pathology , Male , Mice , Nanoparticles/chemistry , Nanoparticles/metabolism , Polyethylene Glycols/chemistry , RAW 264.7 Cells
9.
Biomolecules ; 10(5)2020 05 18.
Article in English | MEDLINE | ID: mdl-32443614

ABSTRACT

A new six intraperitoneal injections insulin-mimetic vanadyl(IV) compounds [(VO)(FA)(AAn)] (where n = 1-6: AA1 = isoleucine, AA2 = threonine, AA3 = proline, AA4 = phenylalanine, AA5 = lysine, and AA6 = glutamine) were synthesized by the chemical reactions between folic acid (FA), VOSO4, and amino acids (AAn) with equal molar ratio 1:1:1 in neutralized media. These complexes were characterized by elemental analysis and estimation of vanadyl(IV) metal ions. The thermal stability behavior of these complexes was studied by TG-DTG-DTA analyses. The structures of these complexes were elucidated by spectroscopic methods like infrared, electron spin resonance (ESR), and solid reflectance spectroscopes. The powder X-ray diffraction (XRD) study suggested the crystalline nature of the complexes. Magnetic moments and electronic spectra revealed the square-pyramid geometrical structure of the complexes. The conductivity results refereed that all synthesized vanadyl(IV) complexes were of a non-electrolyte behavior. The infrared spectra assignments of these complexes revealed that the FAH2 and AAn chelates act as a bidentate ligation. The chelation towards vanadyl (IV) ions existed via deprotonation of one of the carboxylic groups of FAH2 drug ligand, and so amino acids act as bidentate ligands via N-amino and O-carboxylate groups. Both scanning and transmission electron microscope (SEM and TEM) techniques were used to investigate the surface morphology. The main task of this research is the aim of designing a new insulin alternative antidiabetic drug agent. The antidiabetic efficiency of these complexes was evaluated in streptozotocin-induced diabetic male albino rats. Liver and kidney functions, insulin and blood glucose levels, lipid profile, and superoxide dismutase antioxidant (SOD) are verified identifiers for the efficiency of VO(IV)/FA/AAn system compounds as antidiabetic drug agents.


Subject(s)
Biomimetic Materials/chemical synthesis , Coordination Complexes/chemical synthesis , Diabetes Mellitus, Experimental/drug therapy , Folic Acid/analogs & derivatives , Hypoglycemic Agents/chemical synthesis , Insulin/chemistry , Vanadium Compounds/chemistry , Amino Acids/chemistry , Animals , Biomimetic Materials/pharmacokinetics , Biomimetic Materials/therapeutic use , Coordination Complexes/pharmacokinetics , Coordination Complexes/therapeutic use , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/therapeutic use , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Male , Rats
10.
J Nanobiotechnology ; 18(1): 71, 2020 May 13.
Article in English | MEDLINE | ID: mdl-32404183

ABSTRACT

BACKGROUND: Although many therapeutic strategies for Alzheimer's disease (AD) have been explored, these strategies are seldom used in the clinic. Therefore, AD therapeutic research is still urgently needed. One major challenge in the field of nanotherapeutics is to increase the selective delivery of drugs to a targeted location. Herein, we devised and tested a strategy for delivery of nanoparticles to neurons to inhibit tau aggregation by directly targeting p-tau. RESULTS: Curcumin (CUR) is loaded onto red blood cell (RBC) membrane-coated PLGA particles bearing T807 molecules attached to the RBC membrane surface (T807/RPCNP). With the advantage of the suitable physicochemical properties of the PLGA nanoparticles and the unique biological functions of the RBC membrane, the RPCNP are stabilized and promote sustained CUR release, which provided improved biocompatibility and resulted in long-term presence in the circulation. Under the synergistic effects of T807, T807/RPCNP can not only effectively penetrate the blood-brain barrier (BBB), but they also possess high binding affinity to hyperphosphorylated tau in nerve cells where they inhibit multiple key pathways in tau-associated AD pathogenesis. When CUR was encapsulated, our data also demonstrated that CUR-loaded T807/RPCNP NPs can relieve AD symptoms by reducing p-tau levels and suppressing neuronal-like cells death both in vitro and in vivo. The memory impairment observed in an AD mouse model is significantly improved following systemic administration of CUR-loaded T807/RPCNP NPs. CONCLUSION: Intravenous neuronal tau-targeted T807-modified novel biomimetic nanosystems are a promising clinical candidate for the treatment of AD.


Subject(s)
Alzheimer Disease , Biomimetic Materials , Curcumin , Drug Carriers , Nanoparticles/chemistry , Animals , Apoptosis/drug effects , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacokinetics , Blood-Brain Barrier/metabolism , Cell Line , Curcumin/chemistry , Curcumin/pharmacokinetics , Curcumin/pharmacology , Disease Models, Animal , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Hippocampus/drug effects , Hippocampus/pathology , Humans , Maze Learning/drug effects , Mice , Neurons/metabolism , Protective Agents/chemistry , Protective Agents/pharmacokinetics , Protective Agents/pharmacology , tau Proteins/metabolism
11.
Drug Deliv ; 27(1): 502-518, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32228100

ABSTRACT

Reactive oxygen species (ROS)-induced neuronal mitochondrial dysfunction is a key pathologic factor in sporadic Alzheimer's disease (AD). Neuronal mitochondria have been proposed to be a promising therapeutic target for AD, especially for the failures of phase III clinical trials on conventional amyloid-ß (Aß) targeted therapy. However, the efficient intravenous delivery of therapeutic agents to neuronal mitochondria in the brain remains a major challenge due to the complicated physiological environment. Recently, biomaterials-based nanomedicine has been widely investigated for the treatment of AD. Herein, we devised a strategy for functional antioxidant delivery to neuronal mitochondria by loading antioxidants into red blood cell (RBC) membrane-coated nanostructured lipid carriers (NLC) bearing rabies virus glycoprotein (RVG29) and triphenylphosphine cation (TPP) molecules attached to the RBC membrane surface (RVG/TPP NPs@RBCm). With the advantage of suitable physicochemical properties of NLC and unique biological functions of the RBC membrane, RVG/TPP NPs@RBCm are stabilized and enabled sustained drug release, providing improved biocompatibility and long-term circulation. Under the synergistic effects of RVG29 and TPP, RVG/TPP NPs@RBCm can not only penetrate the blood-brain barrier (BBB) but also target neuron cells and further localize in the mitochondria. After encapsulating Resveratrol (RSV) as the model antioxidant, the data demonstrated that RVG/TPP-RSV NPs@RBCm can relieve AD symptoms by mitigating Aß-related mitochondrial oxidative stress both in vitro and in vivo. The memory impairment in APP/PS1 mice is significantly improved following the systemic administration of RVG/TPP-RSV NPs@RBCm. In conclusion, intravenous neuronal mitochondria-targeted dual-modified novel biomimetic nanosystems are a promising therapeutic candidate for ROS-induced mitochondrial dysfunction in AD.


Subject(s)
Alzheimer Disease/drug therapy , Antioxidants/administration & dosage , Biomimetic Materials/chemistry , Mitochondria/drug effects , Nanoparticles/chemistry , Neurons/drug effects , Resveratrol/administration & dosage , Administration, Intravenous , Alzheimer Disease/metabolism , Animals , Antioxidants/pharmacokinetics , Antioxidants/therapeutic use , Biological Transport/drug effects , Biomimetic Materials/pharmacokinetics , Biomimetic Materials/therapeutic use , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/metabolism , Cell Line , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/therapeutic use , Erythrocyte Membrane/chemistry , Male , Mice , Mice, Inbred ICR , Mitochondria/metabolism , Nanoparticles/metabolism , Nanoparticles/therapeutic use , Neurons/metabolism , Organophosphorus Compounds/pharmacokinetics , Organophosphorus Compounds/therapeutic use , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Resveratrol/pharmacokinetics , Resveratrol/therapeutic use , Tissue Distribution
12.
Mater Sci Eng C Mater Biol Appl ; 109: 110566, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32228987

ABSTRACT

Marine demosponges of the Verongiida order are considered a gold-mine for bioinspired materials science and marine pharmacology. The aim of this work was to simultaneously isolate selected bromotyrosines and unique chitinous structures from A. aerophoba and to propose these molecules and biomaterials for possible application as antibacterial and antitumor compounds and as ready-to-use scaffolds for cultivation of cardiomyocytes, respectively. Among the extracted bromotyrosines, the attention has been focused on aeroplysinin-1 that showed interesting unexpected growth inhibition properties for some Gram-negative clinical multi-resistant bacterial strains, such as A. baumannii and K. pneumoniae, and on aeroplysinin-1 and on isofistularin-3 for their anti-tumorigenic activity. For both compounds, the effects are cell line dependent, with significant growth inhibition activity on the neuroblastoma cell line SH-SY5Y by aeroplysinin-1 and on breast cancer cell line MCF-7 by isofistularin-3. In this study, we also compared the cultivation of human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) on the A. aerophoba chitinous scaffolds, in comparison to chitin structures that were pre-coated with Geltrex™, an extracellular matrix mimetic which is used to enhance iPSC-CM adhesion. The iPSC-CMs on uncoated and pure chitin structures started contracting 24 h after seeding, with comparable behaviour observed on Geltrex-coated cell culture plates, confirming the biocompatibility of the sponge biomaterial with this cell type. The advantage of A. aerophoba is that this source organism does not need to be collected in large quantities to supply the necessary amount for further pre-clinical studies before chemical synthesis of the active compounds will be available. A preliminary analysis of marine sponge bioeconomy as a perspective direction for application of biomaterials and secondary bioactive metabolites has been finally performed for the first time.


Subject(s)
Acetonitriles , Alkaloids , Aquatic Organisms/chemistry , Biomimetic Materials , Cyclohexenes , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Porifera/chemistry , Acetonitriles/chemistry , Acetonitriles/pharmacokinetics , Acetonitriles/pharmacology , Alkaloids/chemistry , Alkaloids/pharmacokinetics , Alkaloids/pharmacology , Animals , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacokinetics , Biomimetic Materials/pharmacology , Cell Line, Tumor , Cyclohexenes/chemistry , Cyclohexenes/pharmacokinetics , Cyclohexenes/pharmacology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Humans , Induced Pluripotent Stem Cells/cytology , MCF-7 Cells , Myocytes, Cardiac/cytology
13.
Nat Commun ; 11(1): 1126, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32111847

ABSTRACT

The efficacy of nano-mediated drug delivery has been impeded by multiple biological barriers such as the mononuclear phagocyte system (MPS), as well as vascular and interstitial barriers. To overcome the abovementioned obstacles, we report a nano-pathogenoid (NPN) system that can in situ hitchhike circulating neutrophils and supplement photothermal therapy (PTT). Cloaked with bacteria-secreted outer membrane vesicles inheriting pathogen-associated molecular patterns of native bacteria, NPNs are effectively recognized and internalized by neutrophils. The neutrophils migrate towards inflamed tumors, extravasate across the blood vessels, and penetrate through the tumors. Then NPNs are rapidly released from neutrophils in response to inflammatory stimuli and subsequently taken up by tumor cells to exert anticancer effects. Strikingly, due to the excellent targeting efficacy, cisplatin-loaded NPNs combined with PTT completely eradicate tumors in all treated mice. Such a nano-platform represents an efficient and generalizable strategy towards in situ cell hitchhiking as well as enhanced tumor targeted delivery.


Subject(s)
Chemotaxis, Leukocyte , Drug Delivery Systems , Nanoparticles/administration & dosage , Neoplasms/therapy , Neutrophils/physiology , Phototherapy , Animals , Bacterial Outer Membrane/chemistry , Bacterial Outer Membrane/immunology , Biomimetic Materials/administration & dosage , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacokinetics , Cisplatin/administration & dosage , Cisplatin/chemistry , Cisplatin/pharmacokinetics , Drug Liberation , Extracellular Vesicles/chemistry , Extracellular Vesicles/immunology , Immunotherapy, Adoptive , Inflammation/etiology , Mice , Nanoparticles/chemistry , Nanoparticles/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neutrophil Activation , Neutrophil Infiltration , Neutrophils/immunology , Neutrophils/metabolism , Pathogen-Associated Molecular Pattern Molecules/immunology , Phototherapy/adverse effects , Tumor Microenvironment/radiation effects , Xenograft Model Antitumor Assays
14.
Nano Lett ; 20(3): 1637-1646, 2020 03 11.
Article in English | MEDLINE | ID: mdl-32013452

ABSTRACT

Nanotechnology-based RNA interference (RNAi) has shown great promise in overcoming the limitations of traditional clinical treatments for glioblastoma (GBM). However, because of the complexity of brain physiology, simple blood-brain barrier (BBB) penetration or tumor-targeting strategies cannot entirely meet the demanding requirements of different therapeutic delivery stages. Herein, we developed a charge conversional biomimetic nanoplatform with a three-layer core-shell structure to programmatically overcome persistent obstacles in siRNA delivery to GBM. The resulting nanocomplex presents good biocompatibility, prolonged blood circulation, high BBB transcytosis, effective tumor accumulation, and specific uptake by tumor cells in the brain. Moreover, red blood cell membrane (RBCm) disruption and effective siRNA release can be further triggered elegantly by charge conversion from negative to positive in the endo/lysosome (pH 5.0-6.5) of tumor cells, leading to highly potent target-gene silencing with a strong anti-GBM effect. Our study provides an intelligent biomimetic nanoplatform tailored for systemically siRNA delivery to GBM, leveraging Angiopep-2 peptide-modified, immune-free RBCm and charge conversional components. Improved therapeutic efficacy, higher survival rates, and minimized systemic side effects were achieved in orthotopic U87MG-luc human glioblastoma tumor-bearing nude mice.


Subject(s)
Biomimetic Materials , Brain Neoplasms , Glioblastoma , RNA Interference , RNA, Small Interfering , Animals , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacokinetics , Biomimetic Materials/pharmacology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Mice , Mice, Nude , RNA, Small Interfering/chemistry , RNA, Small Interfering/pharmacokinetics , RNA, Small Interfering/pharmacology , Xenograft Model Antitumor Assays
15.
Nanoscale ; 12(5): 3317-3329, 2020 Feb 07.
Article in English | MEDLINE | ID: mdl-31976511

ABSTRACT

Immune checkpoint blockade (ICB) has emerged as one of the breakthrough approaches for tumor immunotherapy. However, known as an immune "cold" tumor, breast cancer harbors an immunosuppressive tumor niche that compromises ICB-based therapy. Chemoimmunotherapy combines a chemotherapeutic with an immune-modulating agent, representing a promising tactic to combat cancers, while the lack of effectively targeted co-delivery strategy is one of the main obstacles to achieve the synergistic utilization. Herein, self-assembled PEGylated pure drug-based nanohybrids (DNH) were created, which could evoke immunogenic cell death (ICD), aiding ICB-based immunotherapy by controlling the spatiotemporal release of oxaliplatin (OXA) and small molecular inhibitor 1-methyl-d-tryptophan (1-MT). Furthermore, biomimetic functionalization was exploited by nature killer cell membrane camouflaging to target cancerous cells as well as by elicit immune response through inducing M1 macrophage polarization. The drug release profiles of the nanosystem were investigated in the presence of low pH and intracellular reductants. Systemic in vivo bio-behaviors were evaluated via pharmacokinetics and biodistribution. As an "all-in-one" pure drug-based codelivery system, our biomimetic nanoplatform possessed multiple immunomodulation functions, which markedly aided in increasing the frequency of immune responders and generate an immune "hot" breast tumor niche, and eventually allowed to boost breast cancer therapy.


Subject(s)
Biomimetic Materials , Breast Neoplasms , Nanoparticles , Oxaliplatin , Tryptophan/analogs & derivatives , Tumor Microenvironment , Animals , Biomimetic Materials/pharmacokinetics , Biomimetic Materials/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Female , Humans , MCF-7 Cells , Mice, Inbred BALB C , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Oxaliplatin/pharmacokinetics , Oxaliplatin/pharmacology , Tryptophan/pharmacokinetics , Tryptophan/pharmacology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
16.
Biomacromolecules ; 21(2): 688-700, 2020 02 10.
Article in English | MEDLINE | ID: mdl-31769678

ABSTRACT

Bioinspired nonantibiotics can prove to be a better and an efficient tool to fight against antimicrobial resistance. In our study, biomaterial composed of zinc-carboxymethyl chitosan (CMC)-genipin was investigated for this purpose. Briefly, CMC was synthesized and transformed to porous scaffolds using the freeze drying method. The scaffolds were cross-linked and stabilized with genipin and zinc (2 M zinc acetate), respectively. FTIR spectroscopic data testified Zn complex formation and pointed out the absence of water molecule like that of zinc motif containing proteins. Hence, the complex may be termed as biomimetic. Genipin (0.5%) cross-linking appeared to contribute additively to the wet compressive strength of the zinc-CMC scaffolds. Biodegradation data revealed better stability of CMC-genipin-zinc scaffolds in enzymatic and nonenzymatic conditions than their redundant controls. The scaffolds seem to support adhesion and proliferation of human dental pulp stem cells and were hemocompatible to human red blood corpuscles, as revealed by scanning electron microscopy. The scaffolds were found to be antibacterial and mildly antibiofilm when tested against biofilm-forming bacteria, that is, Staphylococcus aureus (ATCC 9144), making it a potential nonantibiotic-like biomaterial. To conclude, this organometallic complex-based biomaterial may potentially serve as a weapon against antimicrobial resistance. Furthermore, the biomaterial potentially finds its application in dental, maxillofacial, and orthopedic tissue engineering applications.


Subject(s)
Adhesives/chemistry , Biocompatible Materials/pharmacokinetics , Biomimetic Materials/pharmacokinetics , Chitosan/analogs & derivatives , Iridoids/chemistry , Zinc/chemistry , Adhesives/pharmacokinetics , Biocompatible Materials/chemistry , Biomimetic Materials/chemistry , Biomimetics/methods , Cells, Cultured , Chitosan/chemistry , Chitosan/pharmacokinetics , Dental Pulp/drug effects , Dental Pulp/metabolism , Erythrocytes/drug effects , Erythrocytes/metabolism , Humans , Iridoids/pharmacokinetics , Materials Testing/methods , Microbial Sensitivity Tests/methods , Tissue Scaffolds , Zinc/pharmacokinetics
17.
Cells ; 8(8)2019 08 13.
Article in English | MEDLINE | ID: mdl-31412631

ABSTRACT

Recently, biomimetic nanoparticles, especially cell membrane-cloaked nanoparticles, have attracted increasing attention in biomedical applications, including antitumor therapy, detoxification, and immune modulation, by imitating the structure and the function of biological systems such as long circulation life in the blood. However, the circulation time of cell membrane-cloaked nanoparticles is far less than that of the original cells, greatly limiting their biomedical applications, while the underlying reasons are seldom demonstrated. In this study, the influence of particle size on the circulation and the biodistribution of red blood cell membrane-coated nanoparticles (RBC-NPs) as model biomimetic nanoparticles were investigated. Differently sized RBC-NPs (80, 120, 160, and 200 nm) were prepared by fusing RBC membranes on poly(lactic-co-glycolic acid) nanoparticles. It was shown that the particle size did not change the cellular uptake of these biomimetic nanoparticles by macrophage cells in vitro and their immunogenic responses in vivo. However, their circulation life in vivo decreased with the particle size, while their accumulation in the liver increased with the particle size, which might be related to their size-dependent filtration through hepatic sinusoids. These findings will provide experimental evidence for the design and the optimization of biomimetic nanoparticles.


Subject(s)
Biomimetic Materials/pharmacokinetics , Coated Materials, Biocompatible/pharmacokinetics , Nanoparticles/chemistry , Particle Size , Animals , Drug Delivery Systems , Erythrocyte Membrane/chemistry , Male , Mice , Mice, Inbred ICR , RAW 264.7 Cells , Tissue Distribution
18.
ACS Appl Mater Interfaces ; 11(34): 30566-30574, 2019 Aug 28.
Article in English | MEDLINE | ID: mdl-31370395

ABSTRACT

The reversible unfolding-refolding transition is considerably important for natural elastomeric proteins (e.g., titin) to fulfill their biological functions. It is of great importance to develop synthetic versions by borrowing their unique stretchable design principles. Herein, we present a novel pulsating vesicle by means of the aqueous self-assembly of supra-amphiphilic helices. Interestingly, this vesicle simultaneously features dynamic swelling and shrinkage movements in response to external proton triggers. Titin-like unfolding-refolding transformation of artificial helices was proved to play a crucial role in this pulsatile motion. Moreover, the vesicular membrane of this vesicle has exhibited tunable permeability during reversible expansion and contraction circulation. Meanwhile, light can also be used as a driving force to further regulate the disassembly-reassembly transformation of the pulsating vesicle. In addition, the drug delivery system was also employed as an investigating model to estimate the permeability variation and disassembly-reassembly behaviors of the pulsating vesicles, which displayed unique dual quick- and sustained-release behaviors toward anti-cancer agents. It is anticipated that this work opens an avenue for fabricating novel stretchable biomimetics by using the exclusive unfolding-refolding nature of artificial foldamers.


Subject(s)
Antineoplastic Agents , Biomimetic Materials , Connectin , Light , Membranes, Artificial , Protein Unfolding , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacokinetics , Biomimetic Materials/pharmacology , Connectin/chemistry , Connectin/pharmacokinetics , Connectin/pharmacology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Humans , Hydrogen-Ion Concentration , MCF-7 Cells , Permeability
19.
Acta Biomater ; 97: 451-460, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31374338

ABSTRACT

Leading causes of vision loss include neovascular age-related macular degeneration (NVAMD) and macular edema (ME), which both require frequent intravitreal injections for treatment. A safe, poly(lactic-co-glycolic acid) (PLGA)-based biodegradable polymeric microparticle (MP) delivery system was developed that encapsulates and protects a biomimetic peptide from degradation, allows sustained intraocular release through polymer hydrolysis, and demonstrates a prolonged anti-angiogenic effect in vivo in three different NVAMD animal models (a laser-induced choroidal neovascularization mouse model, a rhoVEGF transgenic mouse model, and a Tet/opsin/VEGF transgenic mouse model) following intravitreal administration. The role of copolymer composition and microparticle shape was explored and 85:15 lactide-to-glycolide PLGA formed into ellipsoidal microparticles was found to be effective at inhibiting neovascularization for at least 16 weeks in vivo. Treatments were found to not only inhibit the growth of neovascularization, but also to cause regression of the neovasculature, reduce vascular leakage, and prevent exudative retinal detachment. These particulate devices are promising for the sustained release of biologics in the eye and may be useful for treating retinal diseases. STATEMENT OF SIGNIFICANCE: Devastating retinal diseases cause blindness in millions of people around the world. Current protein-based treatments have insufficient efficacy for many patients and also necessitate frequent intravitreal injections. Here, we demonstrate a new treatment consisting of a peptide encapsulated in biodegradable microparticles. We explore the effects of copolymer composition and physical shape of polymeric microparticles and find that both modulate peptide release. Efficacy of the treatment was validated in three different mouse models and the lead formulation was determined to be effective long-term, for at least 16 weeks in vivo, following a single injection. Treatments inhibited and regressed neovascularization as well as reduced vascular leakage. Anisotropic polymeric microparticles are promising for the sustained release of biologics in the eye.


Subject(s)
Biomimetic Materials , Choroidal Neovascularization/prevention & control , Peptides , Polylactic Acid-Polyglycolic Acid Copolymer , Animals , Anisotropy , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacokinetics , Biomimetic Materials/pharmacology , Choroidal Neovascularization/genetics , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Intravitreal Injections , Mice , Mice, Transgenic , Peptides/chemistry , Peptides/pharmacokinetics , Peptides/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology , RAW 264.7 Cells
20.
Acta Biomater ; 96: 517-536, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31284098

ABSTRACT

Exosomes are naturally secreted nanovesicles that have emerged as a promising therapeutic nanodelivery platform, due to their specific composition and biological properties. However, challenges like considerable complexity, low isolation yield, drug payload, and potential safety concerns substantially reduce their pharmaceutical acceptability. Given that the nano-bio-interface is a crucial factor for nanocarrier behavior and function, modification of synthetic nanoparticles with the intrinsic hallmarks of exosomes' membrane to create exosome mimetics could allow for siRNA delivery in a safer and more efficient manner. Herein, connexin 43 (Cx43)-embedded, exosome-mimicking lipid bilayers coated chitosan nanoparticles (Cx43/L/CS NPs) were constructed by using cell-free (CF) synthesis systems with plasmids encoding Cx43 in the presence of lipid-coated CS NPs (L/CS NPs). The integration of de novo synthesized Cx43 into the lipid bilayers of L/CS NPs occurred cotranslationally during one-pot reaction and, more importantly, the integrated Cx43 was functionally active in transport. In addition to considerably lower cytotoxicity (

Subject(s)
Biomimetic Materials , Connexin 43 , Drug Delivery Systems , Exosomes/chemistry , Nanoparticles/chemistry , RNA, Small Interfering , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacokinetics , Biomimetic Materials/pharmacology , Cell-Free System , Connexin 43/biosynthesis , Connexin 43/chemistry , Connexin 43/pharmacokinetics , Connexin 43/pharmacology , HEK293 Cells , Humans , RNA, Small Interfering/chemistry , RNA, Small Interfering/pharmacokinetics , RNA, Small Interfering/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...