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










Publication year range
1.
ACS Appl Mater Interfaces ; 16(19): 25101-25112, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38691046

ABSTRACT

The evolution of nano-drug delivery systems addresses the limitations of conventional cancer treatments with stimulus-responsive nanomaterial-based delivery systems presenting temporal and spatial advantages. Among various nanomaterials, boron nitride nanoparticles (BNNs) demonstrate significant potential in drug delivery and cancer treatment, providing a high drug loading capacity, multifunctionality, and low toxicity. However, the challenge lies in augmenting nanomaterial accumulation exclusively within tumors while preserving healthy tissues. To address this, we introduce a novel approach involving cancer cell membrane-functionalized BNNs (CM-BIDdT) for the codelivery of doxorubicin (Dox) and indocyanine green to treat homologous tumor. The cancer cell membrane biomimetic CM-BIDdT nanoparticles possess highly efficient homologous targeting capabilities toward tumor cells. The surface modification with acylated TAT peptides (dTAT) further enhances the nanoparticle intracellular accumulation. Consequently, CM-BIDdT nanoparticles, responsive to the acidic tumor microenvironment, hydrolyze amide bonds, activate the transmembrane penetrating function, and achieve precise targeting with substantial accumulation at the tumor site. Additionally, the photothermal effect of CM-BIDdT under laser irradiation not only kills cells through thermal ablation but also destroys the membrane on the surface of the nanoparticles, facilitating Dox release. Therefore, the fabricated CM-BIDdT nanoparticles orchestrate chemo-photothermal combination therapy and effectively inhibit tumor growth with minimal adverse effects, holding promise as a new modality for synergistic cancer treatment.


Subject(s)
Boron Compounds , Doxorubicin , Indocyanine Green , Nanoparticles , Doxorubicin/chemistry , Doxorubicin/pharmacology , Indocyanine Green/chemistry , Indocyanine Green/pharmacology , Boron Compounds/chemistry , Boron Compounds/pharmacology , Animals , Humans , Mice , Nanoparticles/chemistry , Cell Line, Tumor , Photothermal Therapy , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , tat Gene Products, Human Immunodeficiency Virus/chemistry , Mice, Inbred BALB C , Drug Carriers/chemistry , Drug Delivery Systems
2.
Behav Brain Res ; 470: 115069, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-38797494

ABSTRACT

Recent clinical studies have highlighted mutations in the voltage-gated potassium channel Kv10.2 encoded by the KCNH5 gene among individuals with autism spectrum disorder (ASD). Our preliminary study found that Kv10.2 was decreased in the hippocampus of valproic acid (VPA) - induced ASD rats. Nevertheless, it is currently unclear how KCNH5 regulates autism-like features, or becomes a new target for autism treatment. We employed KCNH5 knockout (KCNH5-/-) rats and VPA - induced ASD rats in this study. Then, we used behavioral assessments, combined with electrophysiological recordings and hippocampal brain slice, to elucidate the impact of KCNH5 deletion and environmental factors on neural development and function in rats. We found that KCNH5-/- rats showed early developmental delay, neuronal overdevelopment, and abnormal electroencephalogram (EEG) signals, but did not exhibit autism-like behavior. KCNH5-/- rats exposed to VPA (KCNH5-/--VPA) exhibit even more severe autism-like behaviors and abnormal neuronal development. The absence of KCNH5 excessively enhances the activity of the Protein Kinase B (Akt)/Mechanistic Target of Rapamycin (mTOR) signaling pathway in the hippocampus of rats after exposure to VPA. Overall, our findings underscore the deficiency of KCNH5 increases the susceptibility to autism under environmental exposures, suggesting its potential utility as a target for screening and diagnosis in ASD.


Subject(s)
Hippocampus , Proto-Oncogene Proteins c-akt , Signal Transduction , TOR Serine-Threonine Kinases , Animals , TOR Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Hippocampus/metabolism , Hippocampus/drug effects , Rats , Proto-Oncogene Proteins c-akt/metabolism , Male , Neurons/metabolism , Neurons/drug effects , Disease Models, Animal , Valproic Acid/pharmacology , ERG1 Potassium Channel/metabolism , ERG1 Potassium Channel/genetics , Rats, Sprague-Dawley , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/genetics , Autistic Disorder/metabolism , Autistic Disorder/genetics , Ether-A-Go-Go Potassium Channels/metabolism , Ether-A-Go-Go Potassium Channels/genetics
3.
Pharmaceutics ; 15(4)2023 Apr 18.
Article in English | MEDLINE | ID: mdl-37111754

ABSTRACT

Boron nitride nanomaterials are being increasingly recognized as vehicles for cancer drug delivery that increase drug loading and control drug release because of their excellent physicochemical properties and biocompatibility. However, these nanoparticles are often cleared rapidly by the immune system and have poor tumor targeting effects. As a result, biomimetic nanotechnology has emerged to address these challenges in recent times. Cell-derived biomimetic carriers have the characteristics of good biocompatibility, long circulation time, and strong targeting ability. Here, we report a biomimetic nanoplatform (CM@BN/DOX) prepared by encapsulating boron nitride nanoparticles (BN) and doxorubicin (DOX) together using cancer cell membrane (CCM) for targeted drug delivery and tumor therapy. The CM@BN/DOX nanoparticles (NPs) were able to target cancer cells of the same type on its own initiative through homologous targeting of cancer cell membranes. This led to a remarkable increase in cellular uptake. In vitro simulation of an acidic tumor microenvironment could effectively promote drug release from CM@BN/DOX. Furthermore, the CM@BN/DOX complex exhibited an excellent inhibitory effect against homotypic cancer cells. These findings suggest that CM@BN/DOX are promising in targeted drug delivery and potentially personalized therapy against their homologous tumor.

4.
Neuroscience ; 519: 120-130, 2023 05 21.
Article in English | MEDLINE | ID: mdl-36796753

ABSTRACT

The cytoplasmic multifunctional adaptor protein ß-arrestin 2 (Arrb2) is involved in the occurrence of various nervous system diseases, such as Alzheimer's disease and Parkinson's disease. Previous laboratory studies have shown that the expression and function of the Arrb2 gene was increased in valproic acid-induced autistic mice models. However, few reports have examined the possible role of Arrb2 in the pathogenesis of autism spectrum disorder. Therefore, Arrb2-deficient (Arrb2-/-) mice were further studied to uncover the physiological function of Arrb2 in the nervous system. In this study, we found that Arrb2-/- mice had normal behavioral characteristics compared with wild-type mice. The autophagy marker protein LC3B was decreased in the hippocampus of Arrb2-/- mice compared to wild-type mice. Western blot analysis revealed that deletion of Arrb2 caused hyperactivation of Akt-mTOR signaling in the hippocampus. In addition, abnormal mitochondrial dysfunction was observed in Arrb2-/- hippocampal neurons, which was characterized by a reduction in mitochondrial membrane potential and adenosine triphosphate production and an increase in reactive oxygen species levels. Therefore, this study elucidates the interaction between Arrb2 and the Akt-mTOR signaling pathway and provides insights into the role of Arrb2 in hippocampal neuron autophagy.


Subject(s)
Autism Spectrum Disorder , Proto-Oncogene Proteins c-akt , Mice , Animals , beta-Arrestin 1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Autism Spectrum Disorder/pathology , TOR Serine-Threonine Kinases/metabolism , Autophagy/physiology , Hippocampus/metabolism , beta-Arrestin 2/genetics
5.
Nat Commun ; 13(1): 5735, 2022 09 30.
Article in English | MEDLINE | ID: mdl-36175446

ABSTRACT

Theranostic sonosensitizers with combined sonodynamic and near infrared (NIR) imaging modes are required for imaging guided sonodynamic therapy (SDT). It is challenging, however, to realize a single material that is simultaneously endowed with both NIR emitting and sonodynamic activities. Herein, we report the design of a class of NIR-emitting sonosensitizers from a NIR phosphorescent carbon dot (CD) material with a narrow bandgap (1.62 eV) and long-lived excited triplet states (11.4 µs), two of which can enhance SDT as thermodynamically and dynamically favorable factors under low-intensity ultrasound irradiation, respectively. The NIR-phosphorescent CDs are identified as bipolar quantum dots containing both p- and n-type surface functionalization regions that can drive spatial separation of e--h+ pairs and fast transfer to reaction sites. Importantly, the cancer-specific targeting and high-level intratumor enrichment of the theranostic CDs are achieved by cancer cell membrane encapsulation for precision SDT with complete eradication of solid tumors by single injection and single irradiation. These results will open up a promising approach to engineer phosphorescent materials with long-lived triplet excited states for sonodynamic precision tumor therapy.


Subject(s)
Carbon , Quantum Dots , Cell Encapsulation , Cell Membrane , Light
6.
Biochem Biophys Res Commun ; 620: 135-142, 2022 09 10.
Article in English | MEDLINE | ID: mdl-35785569

ABSTRACT

Transient receptor potential vanillic acid 2 (TRPV2) are well recognized for their contributions to neuronal development, cardiac function, immunity and cancer. However, the precise roles for this thermo TRPchannels in neurological disorder remain unknown. In this study, we employed the CRISPR/Cas9 system to generate genetic mutations of TRPV2. Genetic mutation of TRPV2 resulted in autistic-like phenotypes in mice accompanied with the disordered electrical signals recorded by multi-channels in vivo. To determine possible molecular mechanisms, western blotting was further used to assess the possible involvement of several autism-related proteins. The significantly decreased expression of the R2 subunit of the GABA-B receptor in the hippocampus was observed. Together, our findings suggest that genetic mutation of TRPV2 induces autism-like behavior, results in decreased expression of the R2 subunit of the GABA-B receptor.


Subject(s)
Anxiety/genetics , Receptors, GABA-B , TRPV Cation Channels , Animals , Calcium Channels/metabolism , Hippocampus/metabolism , Mice , Mutation , Receptors, GABA-B/metabolism , TRPV Cation Channels/metabolism , gamma-Aminobutyric Acid/metabolism
7.
Am J Cancer Res ; 12(3): 1027-1041, 2022.
Article in English | MEDLINE | ID: mdl-35411249

ABSTRACT

Pancreatic carcinoma (PC) has one of the highest mortality-to-incidence ratios of any solid tumor worldwide. Although KRAS mutation is commonly found in 95% of PCs, directly targeting KRAS remains to be a highly challenging task because of its lacking catalytic pockets where molecule inhibitors can bind with. Proteolysis-targeting chimeric (PROTAC) represents an effective approach for specific degradation of disease-causing proteins by hijacking the endogenous ubiquitin-proteasome system (UPS). Previously, we designed a first-in-class PROTAC induced PDEδ degrader (PIPD), which demonstrated improved anti-tumor efficacy against KRAS mutant malignancies. However, translating cellular degradative effects from bench to beside remains a highly challenging task because of PROTAC's poor penetration efficiency across target cytomembranes and non-targeting delivery induced undesired "off target" side-effects. Herein, a smart nano-drug delivery system (CM8988-PIPD) was successfully constructed by biomimetic strategy for targeted delivery of PIPD. The biomimetic nanoparticle showed well-defined regular spherical structure with an average particle size of approximately 124.8 nm. Cancer cytomembrane camouflage endows CM8988-PIPD with excellent in vivo serum stability, controlled drug release profile, favorable biocompatibility & immunocompatibility, and prominent targeting ability to homologous PC cells. Owing to these advantages, the smart DDS significantly enhanced PDEδ degrading efficacy, resulting in induced cellular apoptosis (more than 50% for both PC cells) and suppressed cell proliferation via the inhibition of RAS signaling. In vitro studies illustrated that CM8988-PIPD hold great potential for the treatment of PC, which merits further investigation in both pre-clinical and clinical investigations in the future.

8.
Pharmaceuticals (Basel) ; 15(2)2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35215270

ABSTRACT

Glioma is among the deadliest types of brain cancer, for which there currently is no effective treatment. Chemotherapy is mainstay in the treatment of glioma. However, drug tolerance, non-targeting, and poor blood-brain barrier penetrance severely inhibits the efficacy of chemotherapeutics. An improved treatment method is thus urgently needed. Herein, a multifunctional biomimetic nanoplatform was developed by encapsulating graphene quantum dots (GQDs) and doxorubicin (DOX) inside a homotypic cancer cell membrane (CCM) for targeted chemo-photothermal therapy of glioma. The GQDs with stable fluorescence and a superior light-to-heat conversion property were synthesized as photothermal therapeutic agents and co-encapsulated with DOX in CCM. The as-prepared nanoplatform exhibited a high DOX loading efficiency. The cell membrane coating protected drugs from leakage. Upon an external laser stimuli, the membrane could be destroyed, resulting in rapid DOX release. By taking advantage of the homologous targeting of the cancer cell membrane, the GQDs/DOX@CCM were found to actively target tumor cells, resulting in significantly enhanced cellular uptake. Moreover, a superior suppression efficiency of GQDs/DOX@CCM to cancer cells through chemo-photothermal treatment was also observed. The results suggest that this biomimetic nanoplatform holds potential for efficient targeting of drug delivery and synergistic chemo-photothermal therapy of glioma.

9.
Int J Nanomedicine ; 16: 2123-2136, 2021.
Article in English | MEDLINE | ID: mdl-33731994

ABSTRACT

PURPOSE: Nanomaterial-based drug-delivery systems allowing for effective targeted delivery of smallmolecule chemodrugs to tumors have revolutionized cancer therapy. Recently, as novel nanomaterials with outstanding physicochemical properties, boron nitride nanospheres (BNs) have emerged as a promising candidate for drug delivery. However, poor dispersity and lack of tumor targeting severely limit further applications. In this study, cancer cell-membrane biomimetic BNs were designed for targeted anticancer drug delivery. METHODS: Cell membrane extracted from HeLa cells (HM) was used to encapsulate BNs by physical extrusion. Doxorubicin (Dox) was loaded onto HM-BNs as a model drug. RESULTS: The cell-membrane coating endowed the BNs with excellent dispersibility and cytocompatibility. The drug-release profile showed that the Dox@HM-BNs responded to acid pH, resulting in rapid Dox release. Enhanced cellular uptake of Dox@HM-BNs by HeLa cells was revealed because of the homologous targeting of cancer-cell membranes. CCK8 and live/dead assays showed that Dox@HM-BNs had stronger cytotoxicity against HeLa cells, due to self-selective cellular uptake. Finally, antitumor investigation using the HeLa tumor model demonstrated that Dox@HM-BNs possessed much more efficient tumor inhibition than free Dox or Dox@BNs. CONCLUSION: These findings indicate that the newly developed HM-BNs are promising as an efficient tumor-selective drug-delivery vehicle for tumor therapy.


Subject(s)
Biomimetic Materials/chemistry , Boron Compounds/chemistry , Cell Membrane/pathology , Molecular Targeted Therapy , Nanospheres/chemistry , Neoplasms/pathology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Body Weight/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Cell Survival/drug effects , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Carriers/chemistry , Drug Liberation , Endocytosis/drug effects , Female , HEK293 Cells , Humans , Mice, Inbred BALB C , Nanospheres/ultrastructure , Neoplasms/drug therapy , Spectrometry, X-Ray Emission , Tissue Distribution/drug effects
10.
Biotech Histochem ; 96(1): 76-81, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32619110

ABSTRACT

Gastric cancer (GC) is characterized by infrequent early diagnosis, poor prognosis and high mortality. Signal transducer and activator of transcription 3 (Stat3) and signal transducer and activator of transcription 5b (Stat5b) play multiple roles in the development and progression of many human cancers. We investigated the effects of silencing Stat3 and Stat5b on the viability and apoptosis of the human gastric cancer cell line, BGC-823. We found that Stat3 and Stat5b were expressed in both the nucleus and cytoplasm of BGC-823 cells. Silencing of Stat3 caused significantly decreased viability and increased apoptosis, as well as attenuated B-cell lymphoma-2 (Bcl-2) expression in BGC-823 cells. Silencing of Stat5b, however, had no significant effect on these events. Stat3, but not Stat5b, plays an important role in the viability and apoptosis of human gastric cancer cell line, BGC-823, which suggests that Stat3 is a potential target for gastric cancer therapy.


Subject(s)
Apoptosis , STAT3 Transcription Factor , Stomach Neoplasms , Cell Line, Tumor , Cell Proliferation , Gene Silencing , Humans , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/genetics
11.
Biomed Res Int ; 2020: 8872577, 2020.
Article in English | MEDLINE | ID: mdl-33204724

ABSTRACT

Autism spectrum disorder (ASD) is a complex neurological disease characterized by impaired social communication and interaction skills, rigid behavior, decreased interest, and repetitive activities. The disease has a high degree of genetic heterogeneity, and the genetic cause of ASD in many autistic individuals is currently unclear. In this study, we report a patient with ASD whose clinical features included social interaction disorder, communication disorder, and repetitive behavior. We examined the patient's genetic variation using whole-exome sequencing technology and found new de novo mutations. After analysis and evaluation, ARRB2 was identified as a candidate gene. To study the potential contribution of the ARRB2 gene to the human brain development and function, we first evaluated the expression profile of this gene in different brain regions and developmental stages. Then, we used weighted gene coexpression network analysis to analyze the associations between ARRB2 and ASD risk genes. Additionally, the spatial conformation and stability of the ARRB2 wild type and mutant proteins were examined by simulations. Then, we further established a mouse model of ASD. The results showed abnormal ARRB2 expression in the mouse ASD model. Our study showed that ARRB2 may be a risk gene for ASD, but the contribution of de novo ARRB2 mutations to ASD is unclear. This information will provide references for the etiology of ASD and aid in the mechanism-based drug development and treatment.


Subject(s)
Autistic Disorder/genetics , Mutation , beta-Arrestin 2/genetics , Animals , Autistic Disorder/chemically induced , Disease Models, Animal , Female , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , Male , Mice, Inbred C57BL , Valproic Acid/toxicity , Exome Sequencing , beta-Arrestin 2/metabolism
12.
Colloids Surf B Biointerfaces ; 190: 110964, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32179413

ABSTRACT

Boron nitride nanospheres (BNNS) have attracted increasing attention in many fields due to their unique physicochemical properties. Biomedical application of BNNS has also been explored recently. However, limited by the hydrophobicity and poor dispersity of BNNS, their biocompatible performance especially the in vivo biosafety has rarely been reported and is still unclear now. In this work, BNNS were firstly camouflaged with red blood cell membrane by physical extrusion (CM-BNNS). CM-BNNS were then incubated with cells as well as intravenously injected into the mice to uncover their potential in vitro and in vivo toxicity. Results were promising as CM-BNNS exhibited better dispersion and stability compared with pristine BNNS. In vitro data demonstrated the relatively enhanced biosafety of CM-BNNS. The red blood cell membrane coating endowed BNNS with markedly prolonged blood circulation and decreased accumulation in the lung. In addition, CM-BNNS showed no adverse effects on all the evaluated hematic parameters and tissues of treated mice at a dose of 10 mg/kg. Taken together, our work demonstrated the optimal biocompatibility of CM-BNNS and pave the way for their future biomedical applications.


Subject(s)
Biocompatible Materials/pharmacology , Boron Compounds/pharmacology , Cell Membrane/chemistry , Erythrocytes/chemistry , Nanospheres/chemistry , Animals , Apoptosis/drug effects , Biocompatible Materials/chemistry , Boron Compounds/chemistry , Cell Survival/drug effects , Cells, Cultured , Drug Carriers/chemistry , Drug Carriers/pharmacology , Drug Delivery Systems , Female , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Inbred Strains , Particle Size , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Surface Properties
13.
Neurochem Res ; 44(12): 2796-2808, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31728858

ABSTRACT

The role of potassium channels provides suggestive evidence for the etiology of autism. The voltage-gated potassium channel Kv10.2 (KCNH5) is widely expressed in the brain. However, the inherent relationship between Kv10.2 and autism is still unclear. Herein, a rat valproic acid (VPA)-induced autism spectrum disorder model was established. The expression level of Kv10.2 was obviously decreased in the hippocampus of VPA rats. Kv10.2 was mainly localized in neurons. Subsequently, a recombinant lentivirus expressing Kv10.2 was used to upregulate the expression of Kv10.2 in the hippocampus of VPA-exposed rats. The results were promising as injection of the Kv10.2 lentivirus in the hippocampus relieved anxiety and stereotypical behavior, and improved the social and exploratory abilities of rats that were prenatally exposed to VPA. In addition, spectral analysis of electroencephalogram data revealed that animals exposed to VPA exhibited increased high-frequency activity compared with the control rats, and this activity recovered to a certain extent after upregulation of Kv10.2 expression by lentivirus injection. These results suggest that changes in Kv10.2 may play an important role in the etiology of autism, thus providing a promising direction for further research on autism.


Subject(s)
Autistic Disorder/therapy , Ether-A-Go-Go Potassium Channels/metabolism , Ether-A-Go-Go Potassium Channels/therapeutic use , Hippocampus/metabolism , Animals , Anxiety/metabolism , Autistic Disorder/chemically induced , Autistic Disorder/etiology , Behavior, Animal/physiology , Biological Therapy , Ether-A-Go-Go Potassium Channels/genetics , Female , Hippocampus/pathology , Lentivirus/genetics , Male , Pregnancy , Rats , Valproic Acid
14.
Neuroreport ; 30(9): 637-644, 2019 06 12.
Article in English | MEDLINE | ID: mdl-31008829

ABSTRACT

Ion channels play as a pivotal role in hypertension in the processes of maintenance of vascular tone and sympathetic excitement of hypertension. The Kv10.2 channel (encoded by the Kcnh5 gene) belongs to the EAG voltage-gated superfamily. It is distributed widely in the brain, such as the hippocampus, the cortex, and the olfactory bulb. To date, the expression of Kv10.2 in central nervous system nuclei that regulates cardiovascular function and its inter-relationship with hypertension are still unclear. Here, electric foot-shock stressors with noise were used to establish the stress-induced hypertensive (SIH) rat model. The expression of Kv10.2 in the rostral ventrolateral medulla, the nucleus tractus solitarius, and the paraventricular nucleus (PVN) was examined by immunohistochemical staining and western blots. The following results were obtained: (a) the expression level of Kv10.2 was increased obviously in the paraventricular nucleus of SIH rats, whereas no significant difference was found in the rostral ventrolateral medulla and the nucleus tractus solitarius. (b) Kv10.2 was located in neurons. (c) Vesicular glutamate transporter 1 as a protein mark of glutamate neurons was increased in the paraventricular nucleus of the SIH group. (d) The expression of vesicular glutamate transporter 1 protein in neurons was significantly decreased when the Kcnh5 gene was knocked down by small interfering RNA in vitro. These findings indicate that the changes in Kv10.2 may be related to SIH, which may provide a potential avenue for further investigation of SIH.


Subject(s)
Brain/metabolism , Ether-A-Go-Go Potassium Channels/biosynthesis , Hypertension/metabolism , Psychological Distress , Animals , Hypertension/etiology , Male , Rats , Rats, Sprague-Dawley
15.
Mater Sci Eng C Mater Biol Appl ; 96: 552-560, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30606565

ABSTRACT

Biomedical application of boron nitride (BN) nanomaterials has recently attracted considerable attentions. BN nanospheres (BNNS) could safely deliver anti-cancer drug into tumor cells, which makes them potential nanocarrier for cancer therapy. However, the poor dispersity in physiological environments and low drug loading capacity severely limit their further applications. Herein, we developed a novel drug delivery system based on folate-conjugated mesoporous silica (MS)-functionalized BNNS (BNMS-FA). Dispersity and drug loading capacity of BNNS were highly improved by MS modification. BNMS-FA complexes were nontoxic up to a concentration of 100 µg/mL, and could be specifically internalized by HeLa and MCF-7 cells via folate receptor-mediated endocytosis. Doxorubicin (DOX) could be loaded onto BNMS-FA complexes with high efficiency via π-π stacking and hydrogen bonding, and showed a sustained release pattern under different pH conditions. BNMS-FA/DOX complexes exhibited superior drug internalization and antitumor efficacy over free DOX, BNNS/DOX and BNMS/DOX complexes, which were considered promising for targeted cancer therapy.


Subject(s)
Boron Compounds , Doxorubicin , Drug Delivery Systems/methods , Folic Acid , Nanospheres , Neoplasms/drug therapy , Silicon Dioxide , Boron Compounds/chemistry , Boron Compounds/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacology , Female , Folic Acid/chemistry , Folic Acid/pharmacology , HeLa Cells , Humans , MCF-7 Cells , Nanospheres/chemistry , Nanospheres/therapeutic use , Neoplasms/metabolism , Neoplasms/pathology , Porosity , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacology
16.
Int J Nanomedicine ; 13: 641-652, 2018.
Article in English | MEDLINE | ID: mdl-29440891

ABSTRACT

BACKGROUND: Anticancer drug-delivery systems (DDSs) capable of responding to the physiological stimuli and efficiently releasing drugs inside tumor cells are highly desirable for effective cancer therapy. Herein, pH-responsive, charge-reversal poly(allylamine hydrochlorid)-citraconic anhydride (PAH-cit) functionalized boron nitride nanospheres (BNNS) were fabricated and used as a carrier for the delivery and controlled release of doxorubicin (DOX) into cancer cells. METHODS: BNNS was synthesized through a chemical vapor deposition method and then functionalized with synthesized charge-reversal PAH-cit polymer. DOX@PAH-cit-BNNS complexes were prepared via step-by-step electrostatic interactions and were fully characterized. The cellular uptake of DOX@PAH-cit-BNNS complexes and DOX release inside cancer cells were visualized by confocal laser scanning microscopy. The in vitro anticancer activity of DOX@ PAH-cit-BNNS was examined using CCK-8 and live/dead viability/cytotoxicity assay. RESULTS: The PAH-cit-BNNS complexes were nontoxic to normal and cancer cells up to a concentration of 100 µg/mL. DOX was loaded on PAH-cit-BNNS complexes with high efficiency. In a neutral environment, the DOX@PAH-cit-BNNS was stable, whereas the loaded DOX was effectively released from these complexes at low pH condition due to amide hydrolysis of PAH-cit. Enhanced cellular uptake of DOX@PAH-cit-BNNS complexes and DOX release in the nucleus of cancer cells were revealed by confocal microscopy. Additionally, the effective delivery and release of DOX into the nucleus of cancer cells led to high therapeutic efficiency. CONCLUSION: Our findings indicated that the newly developed PAH-cit-BNNS complexes are promising as an efficient pH-responsive DDS for cancer therapy.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Doxorubicin/administration & dosage , Drug Delivery Systems/methods , Nanospheres/administration & dosage , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacokinetics , Boron Compounds/chemistry , Citraconic Anhydrides/chemistry , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Drug Liberation , HeLa Cells , Humans , Hydrogen-Ion Concentration , Hydrolysis , MCF-7 Cells , Nanospheres/chemistry , Polyamines/chemistry , Polymers/chemistry , Static Electricity
17.
Nanomaterials (Basel) ; 7(3)2017 Mar 08.
Article in English | MEDLINE | ID: mdl-28336893

ABSTRACT

The application of graphene oxide (GO) as a potential vaccine adjuvant has recently attracted considerable attention. However, appropriate surface functionalization of GO is crucial to improve its biocompatibility and enhance its adjuvant activity. In this study, we developed a simple method to prepare chitosan (CS)-functionalized GO (GO-CS) and further investigated its potential as a nanoadjuvant. Compared with GO, GO-CS possessed considerably smaller size, positive surface charge, and better thermal stability. The functionalization of GO with CS was effective in decreasing the non-specific protein adsorption and improving its biocompatibility. Furthermore, GO-CS significantly activated RAW264.7 cells and stimulated more cytokines for mediating cellular immune response, which was mainly due to the synergistic immunostimulatory effect of both GO and CS. GO-CS exhibits strong potential as a safe nanoadjuvant for vaccines and immunotherapy.

18.
Mater Sci Eng C Mater Biol Appl ; 73: 144-151, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28183591

ABSTRACT

CpG oligodeoxynucleotides (ODNs) activate innate and adaptive immune responses, and show strong potential as immunotherapeutic agents against various diseases. Benefiting from their unique physicochemical properties, graphene oxide (GO) has recently attracted great attention in nanomedicine. In this study, we developed a novel CpG ODNs delivery system based on GO-chitosan (GO-CS) nanocomposites. GO-CS nanocomposites were prepared by self-assembly of both components via electrostatic interactions. Compared with GO, GO-CS nanocomposites possessed smaller size, positive surface charge and lower cytotoxicity. CpG ODNs were loaded onto GO-CS nanocomposites via electrostatic interactions. GO-CS nanocomposites greatly improved the loading capacity and cellular uptake of CpG ODNs. GO-CS/CpG ODNs complexes further resulted in an enhanced interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) production compared with that of free CpG ODNs and GO/CpG ODNs complexes. Therefore, GO-CS nanocomposites can serve as efficient nanocarriers for enhancing the delivery efficiency of CpG ODNs.


Subject(s)
Adjuvants, Immunologic/pharmacology , Chitosan/chemistry , Drug Delivery Systems , Graphite/chemistry , Intracellular Space/metabolism , Nanocomposites/chemistry , Oligodeoxyribonucleotides/pharmacology , Animals , Cell Death/drug effects , Cell Survival/drug effects , Chitosan/chemical synthesis , Cytokines/metabolism , Endocytosis/drug effects , Graphite/chemical synthesis , Mice , Microscopy, Atomic Force , RAW 264.7 Cells , Spectroscopy, Fourier Transform Infrared
19.
Int J Nanomedicine ; 11: 4573-4582, 2016.
Article in English | MEDLINE | ID: mdl-27695318

ABSTRACT

With its unique physical and chemical properties and structural similarity to carbon, boron nitride (BN) has attracted considerable attention and found many applications. Biomedical applications of BN have recently started to emerge, raising great hopes in drug and gene delivery. Here, we developed a targeted anticancer drug delivery system based on folate-conjugated BN nanospheres (BNNS) with receptor-mediated targeting. Folic acid (FA) was successfully grafted onto BNNS via esterification reaction. In vitro cytotoxicity assay showed that BNNS-FA complexes were non-toxic to HeLa cells up to a concentration of 100 µg/mL. Then, doxorubicin hydrochloride (DOX), a commonly used anticancer drug, was loaded onto BNNS-FA complexes. BNNS-FA/DOX complexes were stable at pH 7.4 but effectively released DOX at pH 5.0, which exhibited a pH sensitive and sustained release pattern. BNNS-FA/DOX complexes could be recognized and specifically internalized by HeLa cells via FA receptor-mediated endocytosis. BNNS-FA/DOX complexes exhibited greater cytotoxicity to HeLa cells than free DOX and BNNS/DOX complexes due to the increased cellular uptake of DOX mediated by the FA receptor. Therefore, BNNS-FA complexes had strong potential for targeted cancer therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Boron Compounds/chemistry , Drug Delivery Systems/methods , Nanospheres/administration & dosage , Nanospheres/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Drug Carriers/chemistry , Endocytosis/drug effects , Folic Acid/administration & dosage , Folic Acid/chemistry , HeLa Cells/drug effects , Humans , Spectroscopy, Fourier Transform Infrared
20.
Int J Nanomedicine ; 10: 5343-53, 2015.
Article in English | MEDLINE | ID: mdl-26346655

ABSTRACT

CpG oligodeoxynucleotides (ODNs) stimulate innate and adaptive immune responses. Thus, these molecules are promising therapeutic agents and vaccine adjuvants against various diseases. In this study, we developed a novel CpG ODNs delivery system based on polyethyleneimine (PEI)-functionalized boron nitride nanospheres (BNNS). PEI was coated on the surface of BNNS via electrostatic interactions. The prepared BNNS-PEI complexes had positive zeta potential and exhibited enhanced dispersity and stability in aqueous solution. In vitro cytotoxicity assays revealed that the BNNS-PEI complexes with concentrations up to 100 µg/mL exhibited no obvious cytotoxicity. Furthermore, the positively charged surface of the BNNS-PEI complexes greatly improved the loading capacity and cellular uptake efficiency of CpG ODNs. Class B CpG ODNs loaded on the BNNS-PEI complexes enhanced the production of interleukin-6 and tumor necrosis factor-α from peripheral blood mononuclear cells compared with CpG ODNs directly loaded on BNNS. Contrary to the free CpG ODNs or CpG ODNs directly loaded on BNNS, class B CpG ODNs loaded on the BNNS-PEI complexes induced interferon-α simultaneously. PEI coating may have changed the physical form of class B CpG ODNs on BNNS, which further affected their interaction with Toll-like receptor 9 and induced interferon-α. Therefore, BNNS-PEI complexes can be used to enhance the immunostimulatory effect and therapeutic activity of CpG ODNs and the treatment of diseases requiring interleukin-6, tumor necrosis factor-α, and interferon-α.


Subject(s)
Boron Compounds/chemistry , Drug Carriers , Nanospheres/chemistry , Oligodeoxyribonucleotides/chemistry , Polyethyleneimine/chemistry , Adjuvants, Immunologic/chemistry , Humans , Immune System , Interferon-alpha/metabolism , Interleukin-6/metabolism , Leukocytes, Mononuclear/drug effects , Spectroscopy, Fourier Transform Infrared , Static Electricity , Toll-Like Receptor 9/metabolism , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...