Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nanoscale ; 16(16): 8046-8059, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38563130

ABSTRACT

The biomedical application of nanotechnology in cancer treatment has demonstrated significant potential for improving treatment efficiencies and ameliorating adverse effects. However, the medical translation of nanotechnology-based nanomedicines faces challenges including hazardous environmental effects, difficulties in large-scale production, and possible excessive costs. In the present study, we extracted and purified natural exosome-like nanoparticles (ELNs) from Phellinus linteus. These nanoparticles (denoted as P-ELNs) had an average particle size of 154.1 nm, displayed a negative zeta potential of -31.3 mV, and maintained stability in the gastrointestinal tract. Furthermore, P-ELNs were found to contain a diverse array of functional components, including lipids and pharmacologically active small-molecule constituents. In vitro investigations suggested that they exhibited high internalization efficiency in liver tumor cells (Hepa 1-6) and exerted significant anti-proliferative, anti-migratory, and anti-invasive effects against Hepa 1-6 cells. Strikingly, the therapeutic outcomes of oral P-ELNs were confirmed in an animal model of metastatic hepatocellular carcinoma by amplifying reactive oxygen species (ROS) and rebalancing the gut microbiome. These findings demonstrate the potential of P-ELNs as a promising oral therapeutic platform for liver cancer treatment.


Subject(s)
Carcinoma, Hepatocellular , Exosomes , Gastrointestinal Microbiome , Liver Neoplasms , Reactive Oxygen Species , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Reactive Oxygen Species/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Humans , Mice , Cell Line, Tumor , Exosomes/metabolism , Exosomes/chemistry , Gastrointestinal Microbiome/drug effects , Basidiomycota/chemistry , Basidiomycota/metabolism , Nanoparticles/chemistry , Phellinus/chemistry , Cell Proliferation/drug effects , Cell Movement/drug effects , Administration, Oral
2.
Biomaterials ; 307: 122530, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38493672

ABSTRACT

The therapeutic efficacy of oral nanotherapeutics against colorectal cancer (CRC) is restricted by inadequate drug accumulation, immunosuppressive microenvironment, and intestinal microbiota imbalance. To overcome these challenges, we elaborately constructed 6-gingerol (Gin)-loaded magnetic mesoporous silicon nanoparticles and functionalized their surface with mulberry leaf-extracted lipids (MLLs) and Pluronic F127 (P127). In vitro experiments revealed that P127 functionalization and alternating magnetic fields (AMFs) promoted internalization of the obtained P127-MLL@Gins by colorectal tumor cells and induced their apoptosis/ferroptosis through Gin/ferrous ion-induced oxidative stress and magneto-thermal effect. After oral administration, P127-MLL@Gins safely passed to the colorectal lumen, infiltrated the mucus barrier, and penetrated into the deep tumors under the influence of AMFs. Subsequently, the P127-MLL@Gin (+ AMF) treatment activated antitumor immunity and suppressed tumor growth. We also found that this therapeutic modality significantly increased the abundance of beneficial bacteria (e.g., Bacillus and unclassified-c-Bacilli), reduced the proportions of harmful bacteria (e.g., Bacteroides and Alloprevotella), and increased lipid oxidation metabolites. Strikingly, checkpoint blockers synergistically improved the therapeutic outcomes of P127-MLL@Gins (+ AMF) against orthotopic and distant colorectal tumors and significantly prolonged mouse life spans. Overall, this oral therapeutic platform is a promising modality for synergistic treatment of CRC.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Liposomes , Nanoparticles , Mice , Animals , Colorectal Neoplasms/drug therapy , Nanoparticles/therapeutic use , Administration, Oral , Magnetic Phenomena , Tumor Microenvironment
3.
J Nanobiotechnology ; 22(1): 4, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38169394

ABSTRACT

The clinical application of conventional medications for hepatocellular carcinoma treatment has been severely restricted by their adverse effects and unsatisfactory therapeutic effectiveness. Inspired by the concept of 'medicine food homology', we extracted and purified natural exosome-like lipid nanoparticles (LNPs) from black mulberry (Morus nigra L.) leaves. The obtained MLNPs possessed a desirable hydrodynamic particle size (162.1 nm), a uniform size distribution (polydispersity index = 0.025), and a negative surface charge (-26.6 mv). These natural LNPs were rich in glycolipids, functional proteins, and active small molecules (e.g., rutin and quercetin 3-O-glucoside). In vitro experiments revealed that MLNPs were preferentially internalized by liver tumor cell lines via galactose receptor-mediated endocytosis, increased intracellular oxidative stress, and triggered mitochondrial damage, resulting in suppressing the viability, migration, and invasion of these cells. Importantly, in vivo investigations suggested that oral MLNPs entered into the circulatory system mainly through the jejunum and colon, and they exhibited negligible adverse effects and superior anti-liver tumor outcomes through direct tumor killing and intestinal microbiota modulation. These findings collectively demonstrate the potential of MLNPs as a natural, safe, and robust nanomedicine for oral treatment of hepatocellular carcinoma.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Morus , Nanoparticles , Humans , Carcinoma, Hepatocellular/drug therapy , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Liver Neoplasms/drug therapy , Plant Leaves
4.
ACS Nano ; 18(4): 3651-3668, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38241481

ABSTRACT

Oral administration is the most preferred approach for treating colon diseases, and in situ vaccination has emerged as a promising cancer therapeutic strategy. However, the lack of effective drug delivery platforms hampered the application of in situ vaccination strategy in oral treatment of colorectal cancer (CRC). Here, we construct an oral core-shell nanomedicine by preparing a silk fibroin-based dual sonosensitizer (chlorin e6, Ce6)- and immunoadjuvant (imiquimod, R837)-loaded nanoparticle as the core, with its surface coated with plant-extracted lipids and pluronic F127 (p127). The resultant nanomedicines (Ce6/R837@Lp127NPs) maintain stability during their passage through the gastrointestinal tract and exert improved locomotor activities under ultrasound irradiation, achieving efficient colonic mucus infiltration and specific tumor penetration. Thereafter, Ce6/R837@Lp127NPs induce immunogenic death of colorectal tumor cells by sonodynamic treatment, and the generated neoantigens in the presence of R837 serve as a potent in situ vaccine. By integrating with immune checkpoint blockades, the combined treatment modality inhibits orthotopic tumors, eradicates distant tumors, and modulates intestinal microbiota. As the first oral in situ vaccination, this work spotlights a robust oral nanoplatform for producing a personalized vaccine against CRC.


Subject(s)
Colorectal Neoplasms , Nanoparticles , Vaccines , Humans , Imiquimod , Cell Line, Tumor , Nanomedicine , Colorectal Neoplasms/drug therapy , Vaccination , Immunotherapy
5.
J Mater Chem B ; 12(1): 250-263, 2023 12 22.
Article in English | MEDLINE | ID: mdl-38086697

ABSTRACT

During the wound tissue healing process, the relatively weak driving forces of tissue barriers and concentration gradients lead to a slow and inefficient penetration of bioactive substances into the wound area, consequently showing an impact on the effectiveness of deep wound healing. To overcome these challenges, we constructed biocompatible CaO2-Cu2O "micromotors". These micromotors reacted with the fluids at the wound site, releasing oxygen bubbles and propelling particles deep into the wound tissue. In vitro experimental results revealed that these micromotors not only exhibited antibacterial and hemostatic functions but also facilitated the migration of dermal fibroblasts and vascular endothelial cells, while modulating the inflammatory microenvironment. A methicillin-resistant Staphylococcus aureus infected full-thickness-wound model was created in rats, in which CaO2-Cu2O micromotors markedly expedited the wound healing process. Specifically, CaO2-Cu2O provided a sterile microenvironment for wounds and increased the amounts of M1-type macrophages during infection and inflammation. During the proliferation and remodeling stages, the amount of M1 macrophages gradually decreased, while the amount of M2 macrophages increased, and CaO2-Cu2O did not prolong the inflammatory period. Furthermore, the introduction of a regenerated silk fibroin (RSF) film on the wound surface successfully enhanced the therapeutic effects of CaO2-Cu2O against the infected wound. The combined application of oxygen-producing CaO2-Cu2O micromotors and a RSF film demonstrates significant therapeutic potential and emerges as a promising candidate for the treatment of infected wounds.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Rats , Animals , Endothelial Cells , Wound Healing , Anti-Bacterial Agents/pharmacology , Cell Movement , Hemostasis , Oxygen/pharmacology
6.
Biomaterials ; 302: 122332, 2023 11.
Article in English | MEDLINE | ID: mdl-37801790

ABSTRACT

The treatment efficacies of conventional medications against colorectal cancer (CRC) are restricted by a low penetrative, hypoxic, and immunosuppressive tumor microenvironment. To address these restrictions, we developed an innovative antitumor platform that employs calcium overload-phototherapy using mitochondrial N770-conjugated mesoporous silica nanoparticles loaded with CaO2 (CaO2-N770@MSNs). A loading level of 14.0 wt% for CaO2-N770@MSNs was measured, constituting an adequate therapeutic dosage. With the combination of oxygen generated from CaO2 and hyperthermia under near-infrared irradiation, CaO2-N770@MSNs penetrated through the dense mucus, accumulated in the colorectal tumor tissues, and inhibited tumor cell growth through endoplasmic reticulum stress and mitochondrial damage. The combination of calcium overload and phototherapy revealed high therapeutic efficacy against orthotopic colorectal tumors, alleviated the immunosuppressive microenvironment, elevated the abundance of beneficial microorganisms (e.g., Lactobacillaceae and Lachnospiraceae), and decreased harmful microorganisms (e.g., Bacteroidaceae and Muribaculaceae). Moreover, together with immune checkpoint blocker (αPD-L1), these nanoparticles showed an ability to eradicate both orthotopic and distant tumors, while potentiating systemic antitumor immunity. This treatment platform (CaO2-N770@MSNs plus αPD-L1) open a new horizon of synergistic treatment against hypoxic CRC with high killing power and safety.


Subject(s)
Colorectal Neoplasms , Hyperthermia, Induced , Nanoparticles , Humans , Calcium , Cell Line, Tumor , Phototherapy , Colorectal Neoplasms/therapy , Immunotherapy , Hypoxia , Tumor Microenvironment
7.
J Nanobiotechnology ; 21(1): 6, 2023 Jan 04.
Article in English | MEDLINE | ID: mdl-36600299

ABSTRACT

While several artificial nanodrugs have been approved for clinical treatment of breast tumor, their long-term applications are restricted by unsatisfactory therapeutic outcomes, side reactions and high costs. Conversely, edible plant-derived natural nanotherapeutics (NTs) are source-widespread and cost-effective, which have been shown remarkably effective in disease treatment. Herein, we extracted and purified exosome-like NTs from tea leaves (TLNTs), which had an average diameter of 166.9 nm and a negative-charged surface of - 28.8 mV. These TLNTs contained an adequate slew of functional components such as lipids, proteins and pharmacologically active molecules. In vitro studies indicated that TLNTs were effectively internalized by breast tumor cells (4T1 cells) and caused a 2.5-fold increase in the amount of intracellular reactive oxygen species (ROS) after incubation for 8 h. The high levels of ROS triggered mitochondrial damages and arrested cell cycles, resulting in the apoptosis of tumor cells. The mouse experiments revealed that TLNTs achieved good therapeutic effects against breast tumors regardless of intravenous injection and oral administration through direct pro-apoptosis and microbiota modulation. Strikingly, the intravenous injection of TLNTs, not oral administration, yielded obvious hepatorenal toxicity and immune activation. These findings collectively demonstrate that TLNTs can be developed as a promising oral therapeutic platform for the treatment of breast cancer.


Subject(s)
Exosomes , Mammary Neoplasms, Animal , Microbiota , Animals , Mice , Exosomes/metabolism , Reactive Oxygen Species/metabolism , Mammary Neoplasms, Animal/pathology , Apoptosis , Plant Leaves/metabolism , Tea , Cell Line, Tumor
8.
Small ; 18(42): e2203466, 2022 10.
Article in English | MEDLINE | ID: mdl-36117129

ABSTRACT

The therapeutic outcomes of oral nanomedicines against colon cancer are heavily compromised by their lack of specific penetration into the internal tumor, favorable anti-tumor activity, and activation of anti-tumor immunity. Herein, hydrogen peroxide (H2 O2 )/ultrasound (US)-driven mesoporous manganese oxide (MnOx )-based nanomotors are constructed by loading mitochondrial sonosensitizers into their mesoporous channels and orderly dual-functionalizing their surface with silk fibroin and chondroitin sulfate. The locomotory activities and tumor-targeting capacities of the resultant nanomotors (CS-ID@NMs) are greatly improved in the presence of H2 O2 and US irradiation, inducing efficient mucus-traversing and deep tumor penetration. The excess H2 O2 in the tumor microenvironment (TME) is decomposed into hydroxyl radicals and oxygen by an Mn2+ -mediated Fenton-like reaction, and the produced oxygen participates in sonodynamic therapy (SDT), yielding abundant singlet oxygen. The combined Mn2+ -mediated chemodynamic therapy and SDT cause effective ferropotosis of tumor cells and accelerate the release of tumor antigens. Importantly, animal experiments reveal that the treatment of combining oral hydrogel (chitosan/alginate)-embedding CS-ID@NMs and immune checkpoint inhibitors can simultaneously suppress the growth of primary and distal tumors through direct killing, reversion of immunosuppressive TME, and potentiation of systemic anti-tumor immunity, demonstrating that the CS-ID@NM-based platform is a robust oral system for synergistic treatment of colon cancer.


Subject(s)
Chitosan , Colonic Neoplasms , Fibroins , Nanoparticles , Neoplasms , Animals , Singlet Oxygen/pharmacology , Chitosan/pharmacology , Hydrogen Peroxide/pharmacology , Chondroitin Sulfates/pharmacology , Chondroitin Sulfates/therapeutic use , Cell Line, Tumor , Immune Checkpoint Inhibitors , Neoplasms/therapy , Colonic Neoplasms/drug therapy , Tumor Microenvironment , Oxygen/pharmacology , Mucus , Antigens, Neoplasm , Hydrogels/pharmacology , Alginates , Nanoparticles/therapeutic use
9.
Acta Pharm Sin B ; 12(2): 907-923, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35256954

ABSTRACT

Although several artificial nanotherapeutics have been approved for practical treatment of metastatic breast cancer, their inefficient therapeutic outcomes, serious adverse effects, and high cost of mass production remain crucial challenges. Herein, we developed an alternative strategy to specifically trigger apoptosis of breast tumors and inhibit their lung metastasis by using natural nanovehicles from tea flowers (TFENs). These nanovehicles had desirable particle sizes (131 nm), exosome-like morphology, and negative zeta potentials. Furthermore, TFENs were found to contain large amounts of polyphenols, flavonoids, functional proteins, and lipids. Cell experiments revealed that TFENs showed strong cytotoxicities against cancer cells due to the stimulation of reactive oxygen species (ROS) amplification. The increased intracellular ROS amounts could not only trigger mitochondrial damage, but also arrest cell cycle, resulting in the in vitro anti-proliferation, anti-migration, and anti-invasion activities against breast cancer cells. Further mice investigations demonstrated that TFENs after intravenous (i.v.) injection or oral administration could accumulate in breast tumors and lung metastatic sites, inhibit the growth and metastasis of breast cancer, and modulate gut microbiota. This study brings new insights to the green production of natural exosome-like nanoplatform for the inhibition of breast cancer and its lung metastasis via i.v. and oral routes.

10.
Acta Pharm Sin B ; 12(1): 406-423, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35127395

ABSTRACT

Incorporation of multiple functions into one nanoplatform can improve cancer diagnostic efficacy and enhance anti-cancer outcomes. Here, we constructed doxorubicin (DOX)-loaded silk fibroin-based nanoparticles (NPs) with surface functionalization by photosensitizer (N770). The obtained nanotheranostics (N770-DOX@NPs) had desirable particle size (157 nm) and negative surface charge (-25 mV). These NPs presented excellent oxygen-generating capacity and responded to a quadruple of stimuli (acidic solution, reactive oxygen species, glutathione, and hyperthermia). Surface functionalization of DOX@NPs with N770 could endow them with active internalization by cancerous cell lines, but not by normal cells. Furthermore, the intracellular NPs were found to be preferentially retained in mitochondria, which were also efficient for near-infrared (NIR) fluorescence imaging, photothermal imaging, and photoacoustic imaging. Meanwhile, DOX could spontaneously accumulate in the nucleus. Importantly, a mouse test group treated with N770-DOX@NPs plus NIR irradiation achieved the best tumor retardation effect among all treatment groups based on tumor-bearing mouse models and a patient-derived xenograft model, demonstrating the unprecedented therapeutic effects of trimodal imaging-guided mitochondrial phototherapy (photothermal therapy and photodynamic therapy) and chemotherapy. Therefore, the present study brings new insight into the exploitation of an easy-to-use, versatile, and robust nanoplatform for programmable targeting, imaging, and applying synergistic therapy to tumors.

11.
Biomaterials ; 279: 121178, 2021 12.
Article in English | MEDLINE | ID: mdl-34656857

ABSTRACT

Although synthesized nanotherapeutics (NTs) are attractive for the oral treatment of colon diseases, their clinical translations are constrained by the unsatisfactory therapeutic outcomes, potential adverse effects, and high cost of mass production. Here, we report the development of tea leaf-derived natural NTs with desirable particle sizes (140.0 nm) and negative surface charge (-14.6 mV). These natural exosome-like NTs were found to contain large amounts of lipids, some functional proteins, and many bioactive small molecules. Specifically, galactose groups on the surface of NTs could mediate their specific internalization by macrophages via galactose receptor-mediated endocytosis. Moreover, these NTs were able to reduce the production of reactive oxygen species, inhibit the expression of pro-inflammatory cytokines, and increase the amount of anti-inflammatory IL-10 secreted by macrophages. Orally administered NTs could efficiently inhibit the inflammatory bowel responses, restore disrupted colonic barriers and enhance the diversity and overall abundance of gut microbiota, thereby preventing or alleviating inflammatory bowel disease and colitis-associated colon cancer. The present study brings new insights to the facile application of a versatile and robust natural nanoplatform for the prevention and treatment of colon diseases.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Animals , Colitis/drug therapy , Colitis/prevention & control , Colon , Cytokines , Disease Models, Animal , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/prevention & control , Plant Leaves , Tea
12.
Adv Drug Deliv Rev ; 176: 113887, 2021 09.
Article in English | MEDLINE | ID: mdl-34314785

ABSTRACT

The incidence of inflammatory bowel disease (IBD) is rapidly rising throughout the world. Although tremendous efforts have been made, limited therapeutics are available for IBD management. Natural active small molecules (NASMs), which are a gift of nature to humanity, have been widely used in the prevention and alleviation of IBD; they have numerous advantageous features, including excellent biocompatibility, pharmacological activity, and mass production potential. Oral route is the most common and acceptable approach for drug administration, but the clinical application of NASMs in IBD treatment via oral route has been seriously restricted by their inherent limitations such as high hydrophobicity, instability, and poor bioavailability. With the development of nanotechnology, polymeric nanoparticles (NPs) have provided a promising platform that can efficiently encapsulate versatile NASMs, overcome multiple drug delivery barriers, and orally deliver the loaded NASMs to targeted tissues or cells while enhancing their stability and bioavailability. Thus, NPs can enhance the preventive and therapeutic effects of NASMs against IBD. Herein, we summarize the recent knowledge about polymeric matrix-based carriers, targeting ligands for drug delivery, and NASMs. We also discuss the current challenges and future developmental directions.


Subject(s)
Biological Products/administration & dosage , Inflammatory Bowel Diseases/drug therapy , Nanoparticles/administration & dosage , Polymers/administration & dosage , Administration, Oral , Animals , Humans , Ligands
13.
ACS Appl Mater Interfaces ; 12(45): 50896-50908, 2020 Nov 11.
Article in English | MEDLINE | ID: mdl-33107728

ABSTRACT

It remains a great challenge to design a multifunctional and robust nanoplatform for stimuli-responsive drug delivery toward a lesion, which tactfully integrates multiple molecules with therapeutic and diagnostic characteristics. Herein, we reported a facile and ingenious cross-linked nanogel (DSA) based on the chemical cross-link of drugs as a straightforward strategy to overcome the instability of the assembly. In DSA, doxorubicin (DOX) and 5-aminolevulinic acid (ALA) were cross-linked with a disulfide linker for realizing synergistic anticancer therapy. The stability of DSA was adjusted via balancing the hydrophobic/hydrophilic property with hydrophilic NH2-PEG1k. After regulating the coordination of the DOX part and ALA moiety, the drug-loaded nanogel exhibited superior chemotherapeutic efficacies. Additionally, the DSA could selectively biosynthesize fluorescent protoporphyrin IX (PpIX) in tumor cells, which could be applied for a real-time imaging probe of accurate cancer diagnosis. Besides, the in situ synthesized PpIX in mitochondria could serve as a photosensitizer to convert oxygen into toxic reactive oxygen species under a near infrared ray at 660 nm irradiation, leading to an excellent tumor-killing efficacy. This work proposed a unique strategy for designing a series of prodrug nanogels as a universal drug delivery platform for realizing precise disease therapy and diagnostics.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Glutathione/metabolism , Levulinic Acids/pharmacology , Nanogels/chemistry , Photochemotherapy , Theranostic Nanomedicine , Animals , Antibiotics, Antineoplastic/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Doxorubicin/chemistry , Drug Screening Assays, Antitumor , Female , Glutathione/analysis , Hydrophobic and Hydrophilic Interactions , Levulinic Acids/chemistry , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Molecular Structure , Particle Size , Rats , Rats, Sprague-Dawley , Surface Properties , Aminolevulinic Acid
14.
J Control Release ; 327: 371-383, 2020 11 10.
Article in English | MEDLINE | ID: mdl-32810527

ABSTRACT

The integration of multimodal functions into one nanoplatform holds great promise for enhancing anticancer drug action and mitigating adverse effects. Herein, we prepared hyaluronic acid-functionalized regenerated silk fibroin-based nanoparticles (NPs) loading with photosensitizer (NIR770) and doxorubicin (DOX). The resultant HNDNPs had a desirable diameter of 161.0 nm and a negative zeta-potential of -30.5 mV. Interestingly, they showed excellent responses when triggered with various stimuli (acidity, reactive oxygen species, glutathione, hyaluronidase, or hyperthermia). Cell experiments revealed that HNDNPs could be specifically internalized by A549 cells, and efficiently released the payloads into the cytoplasm. Moreover, NIR770 was preferentially retained in mitochondria due to its lipophilic and cationic properties, which exhibited highly efficient photothermal therapy and photodynamic therapy upon near infrared (NIR) irradiation. Meanwhile, DOX molecules were mainly accumulated in the nucleus. Intravenous injection of HNDNPs into mice followed by NIR irradiation provided excellent multimodal imaging (NIR, photothermal, and photoacoustic imaging), almost eliminated the entire tumor, and greatly prolonged mice survival time with no side effects. Our study demonstrates that this HNDNP, which integrates the functions of tumor targeting, on-demand drug release, multimodal imaging, mitochondrial phototherapy, and chemotherapy, can be exploited as a promising nanococktail for imaging-guided synergistic treatment of cancer.


Subject(s)
Hyperthermia, Induced , Nanoparticles , Photochemotherapy , Animals , Cell Line, Tumor , Doxorubicin , Mice , Mice, Inbred BALB C , Mitochondria , Phototherapy
15.
Colloids Surf B Biointerfaces ; 187: 110747, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31924469

ABSTRACT

Edible plant-derived nanoparticles (NPs) have attracted increasing attention in the treatment of ulcerative colitis (UC). Lycium barbarum (LB), a popular functional fruit, possesses various biological functions. Here, fat-soluble contents were extracted from LB and further processed into LB lipid-derived NPs (LBLNs). The resultant NPs had an average hydrodynamic diameter around 189.2 nm, narrow size distribution (polydispersity index = 0.2), and negative surface charge (-34.9 mV). Moreover, they could be efficiently taken up by UC therapy-related target cells (macrophages), and over 69.0 % of macrophages internalized LBLNs after 4 h co-incubation. We further found from the in vitro results that LBLNs had strong capacities to inhibit the secretion of the main pro-inflammatory cytokines (TNF-α and IL-12) and up-regulate the expression of the typical anti-inflammatory factor (IL-10). Finally, mice experiments confirmed that LBLNs after oral administration could specifically accumulate into inflamed colon tissues, and further attenuate UC-relevant symptoms (e.g., bodyweight loss, colon shortening, increase of spleen weight, and histopathological appearance, as well as ulceration). Collectively, this study demonstrates the excellent therapeutic outcomes of LBLNs against UC and provides a promising edible nanotherapeutic for UC treatment.


Subject(s)
Colitis, Ulcerative/drug therapy , Colon/drug effects , Drug Carriers/administration & dosage , Lycium/chemistry , Macrophages/drug effects , Nanoparticles/administration & dosage , Administration, Oral , Animals , Colitis, Ulcerative/blood , Colitis, Ulcerative/physiopathology , Colon/cytology , Colon/metabolism , Colon/physiopathology , Drug Carriers/chemistry , Drug Delivery Systems/methods , Flavonoids/analysis , Heart/drug effects , Heart/physiology , Interleukin-10/metabolism , Interleukin-12/metabolism , Kidney/drug effects , Kidney/metabolism , Lipids/chemistry , Liver/drug effects , Liver/metabolism , Lung/drug effects , Lung/metabolism , Macrophages/metabolism , Male , Mice , Nanoparticles/chemistry , Particle Size , RAW 264.7 Cells , Spleen/drug effects , Spleen/metabolism , Tumor Necrosis Factor-alpha/metabolism
16.
Mater Sci Eng C Mater Biol Appl ; 106: 110253, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31753332

ABSTRACT

The application of photoresponsive surface molecularly imprinted polymers based on azobenzene is limited by the UV light source required and their poor water solubility. Reducing the phototoxicity and solvent toxicity of the polymers therefore presents a challenge. In this work, an NIR-light-responsive surface molecularly imprinted polymer was fabricated by atom transfer radical polymerization using up-conversion nanoparticles as the core, a hydrophilic green-light-responsive azobenzene derivative as the functional monomer, and a drug as the template. The up-conversion nanoparticles core emitted green fluorescence in the range of 520-550 nm upon NIR irradiation (980 nm, 5 W cm-2), which was absorbed by the azobenzene containing molecularly imprinted polymers layer on the up-conversion nanoparticles surface. This caused the azobenzene chromophores to undergo trans→cis isomerization in phosphate buffered solution (pH = 7.4), thus resulting in NIR-light-induced drug release. The up-conversion fluorescence spectra were used to study the interaction mechanism between the azobenzene monomer and NIR light. Compared with structural analogues of the template (antifebrin and phenacetin), the NIR-light-responsive surface molecularly imprinted polymer showed excellent specificity of recognition for the template drug (paracetamol). The maximum adsorption capacity of the NIR-light-responsive surface molecularly imprinted polymer for loading of paracetamol was 16.80 µmol g-1. The NIR-light-responsive surface molecularly imprinted polymer was applied for NIR-light-induced paracetamol release in phosphate buffered solution (pH = 7.4) through porcine tissue. This work demonstrates the potential of drug delivery systems based on molecularly imprinted polymers for application in deep tissue delivery.


Subject(s)
Molecular Imprinting/methods , Polymers/chemistry , Animals , Azo Compounds/chemistry , Cell Line, Tumor , Drug Liberation , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Microscopy, Electrochemical, Scanning , Polymerization , Swine
17.
Carbohydr Polym ; 223: 115126, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31426992

ABSTRACT

Orally targeted delivery of anti-inflammatory drugs to macrophages has attracted great attention for minimizing the symptoms of ulcerative colitis (UC). In this investigation, we encapsulated curcumin (CUR) into polymeric nanoparticles (NPs), and conjugated chondroitin sulfate (CS) to their surfaces. The resulting CS-NPs had an average diameter of 281 nm, monodisperse size distribution and negatively charged surface. Cell experiments indicated that these NPs showed excellent biocompatibility, and yielded significantly higher cell internalization efficiency in Raw 264.7 macrophages than their counterparts (carboxymethyl cellulose-functionalized CUR-encapsulated NPs, CUL-NPs). Moreover, CS-NPs exhibited a dramatically stronger capacity to inhibit the secretion of the major pro-inflammatory cytokines from lipopolysaccharide-stimulated macrophages compared with CUL-NPs. In vivo experiments revealed that oral administration of chitosan/alginate hydrogel embedding CS-NPs achieved better therapeutic outcomes against UC comparied with CUL-NPs. Collectively, our results demonstrated that CS-NP-embedded hydrogel held a great promise to be developed as a macrophage-targeted drug delivery system for UC treatment.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Chondroitin Sulfates/chemistry , Colitis, Ulcerative/drug therapy , Colon/drug effects , Curcumin/pharmacology , Drug Delivery Systems , Macrophages/drug effects , Nanoparticles/chemistry , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Chondroitin Sulfates/administration & dosage , Chondroitin Sulfates/pharmacology , Colitis, Ulcerative/metabolism , Colon/metabolism , Curcumin/administration & dosage , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , Nanoparticles/administration & dosage , Particle Size , RAW 264.7 Cells , Surface Properties
18.
ACS Appl Mater Interfaces ; 11(32): 29330-29340, 2019 Aug 14.
Article in English | MEDLINE | ID: mdl-31329411

ABSTRACT

Mitochondria-targeting cancer therapies have achieved unprecedented advances attributed to their superior ability for improving drug delivery efficiency and producing an enhanced therapeutic effect. Herein, we report a mitochondria-targeting camptothecin (CPT) polyprodrug system (MCPS) covalently decorated with a high-proportioned CPT content, which can realize drug release specifically responsive to a tumor microenvironment. The nonlinear structure of MCPS can form water-soluble unimolecular micelles with high micellar stability and improved drug accumulation in tumoral cells/tissues. Furthermore, a classical mitochondria-targeting agent, triphenylphosphonium bromide, was tethered in this prodrug system, which causes mitochondrial membrane potential depolarization and mediates the transport of CPT into mitochondria. The disulfide bond in MCPS can be cleaved by an intracellular reductant such as glutathione, leading to enhanced destruction of mitochondria DNA and cell apoptosis induced by a high level of reactive oxygen species. The systematic analyses both in vitro and in vivo indicated the excellent tumor inhibition effect and biosafety of MCPS, which is believed to be an advantageous nanoplatform for subcellular organelle-specific chemotherapy of cancer.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Camptothecin , Mammary Neoplasms, Experimental , Mitochondria , Prodrugs , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Camptothecin/chemistry , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Female , HeLa Cells , Humans , MCF-7 Cells , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mice, Inbred BALB C , Micelles , Mitochondria/metabolism , Mitochondria/pathology , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Reactive Oxygen Species/metabolism
19.
Colloids Surf B Biointerfaces ; 177: 399-406, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30785037

ABSTRACT

Targeted delivery of chemotherapeutic drugs to tumors is a major challenge in colon cancer chemotherapy. To overcome this bottleneck, we loaded camptothecin (CPT) into polymeric nanoparticles (NPs), and further functionalized their surface with chondroitin sulfate (CS). The resulting CS-CPT-NPs had a desirable hydrodynamic diameter (289 nm), narrow particle size distribution (polydispersity index = 0.192) and neutral surface charge. Furthermore, in vitro experiments revealed that the surface functionalization of CS endowed NPs with the capacity of colon cancer-targeted drug delivery, and significantly improved the anti-colon cancer activities and pro-apoptosis effects against colon cancer cells. Strikingly, treatment of colon tumor-bearing mice with different NPs clearly indicated that CS-CPT-NPs showed much better therapeutic outcomes than non-targeted NPs and no systemic toxicity. Taken together, these results demonstrated the promising potential of CS-CPT-NP as an effective drug delivery system for colon cancer-targeted chemotherapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/pharmacology , Chondroitin Sulfates/chemistry , Colonic Neoplasms/drug therapy , Nanoparticles/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology , Animals , Antineoplastic Agents, Phytogenic/chemistry , Apoptosis/drug effects , Camptothecin/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Colonic Neoplasms/pathology , Drug Delivery Systems , Drug Screening Assays, Antitumor , Humans , Hydrodynamics , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Surface Properties , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...