Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 139
Filter
1.
Hepatology ; 78(1): 88-102, 2023 07 01.
Article in English | MEDLINE | ID: mdl-36947402

ABSTRACT

BACKGROUND AND AIMS: Gut microbiota are recognized to be important for anticancer therapy, yet the underlying mechanism is not clear. Here, through the analysis of clinical samples, we identify the mechanism by which the gut microbial metabolite butyrate inhibits HCC and then explore new strategies for HCC treatment. APPROACH AND RESULTS: In our study, we demonstrate that gut microbial metabolite butyrate improves anticancer therapy efficacy by regulating intracellular calcium homeostasis. Using liquid chromatography-mass spectrometry analysis, we found that butyrate metabolism is activated in HCC patients compared with healthy individuals. Butyrate levels are lower in the plasma of HCC patients by gas chromatography-mass spectrometry (GC-MS) analysis. Butyrate supplementation or depletion of short-chain Acyl-CoA dehydrogenase (SCAD) gene (ACADS), encoding a key enzyme for butyrate metabolism, significantly inhibits HCC proliferation and metastasis. The profiling analysis of genes upregulated by butyrate supplementation or ACADS knockdown reveals that calcium signaling pathway is activated, leading to dysregulation of intracellular calcium homeostasis and production of reactive oxygen species. Butyrate supplementation improves the therapy efficacy of a tyrosine kinase inhibitor sorafenib. On the basis of these findings, we developed butyrate and sorafenib coencapsulated mPEG-PLGA-PLL nanoparticles coated with anti-GPC3 antibody (BS@PEAL-GPC3) to prolong the retention time of drugs and enhance drug targeting, leading to high anticancer efficacy. BS@PEAL-GPC3 nanoparticles significantly reduce HCC progression. In addition, BS@PEAL-GPC3 nanoparticles display excellent HCC targeting with excellent safety. CONCLUSIONS: In conclusion, our findings provide new insight into the mechanism by which the gut microbial metabolites inhibit HCC progression, suggesting a translatable therapeutics approach to enhance the clinical targeted therapeutic efficacy.


Subject(s)
Antineoplastic Agents , Butyrates , Carcinoma, Hepatocellular , Gastrointestinal Microbiome , Liver Neoplasms , Sorafenib , Butyrates/pharmacology , Calcium/metabolism , Carcinoma, Hepatocellular/drug therapy , Homeostasis , Liver Neoplasms/drug therapy , Sorafenib/therapeutic use , Antineoplastic Agents/therapeutic use
2.
J Nanobiotechnology ; 22(1): 377, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937768

ABSTRACT

BACKGROUND: Efficient monitoring of glucose concentration in the human body necessitates the utilization of electrochemically active sensing materials in nonenzymatic glucose sensors. However, prevailing limitations such as intricate fabrication processes, lower sensitivity, and instability impede their practical application. Herein, ternary Cu-Co-Ni-S sulfides nanoporous network structure was synthesized on carbon fiber paper (CP) by an ultrafast, facile, and controllable technique through on-step cyclic voltammetry, serving as a superior self-supporting catalytic electrode for the high-performance glucose sensor. RESULTS: The direct growth of free-standing Cu-Co-Ni-S on the interconnected three-dimensional (3D) network of CP boosted the active site of the composites, improved ion diffusion kinetics, and significantly promoted the electron transfer rate. The multiple oxidation states and synergistic effects among Co, Ni, Cu, and S further promoted glucose electrooxidation. The well-architected Cu-Co-Ni-S/CP presented exceptional electrocatalytic properties for glucose with satisfied linearity of a broad range from 0.3 to 16,000 µM and high sensitivity of 6829 µA mM- 1 cm- 2. Furthermore, the novel sensor demonstrated excellent selectivity and storage stability, which could successfully evaluate the glucose levels in human serum. Notably, the novel Cu-Co-Ni-S/CP showed favorable biocompatibility, proving its potential for in vivo glucose monitoring. CONCLUSION: The proposed 3D hierarchical morphology self-supported electrode sensor, which demonstrates appealing analysis behavior for glucose electrooxidation, holds great promise for the next generation of high-performance glucose sensors.


Subject(s)
Biosensing Techniques , Carbon Fiber , Cobalt , Copper , Electrochemical Techniques , Electrodes , Nickel , Sulfides , Copper/chemistry , Nickel/chemistry , Catalysis , Humans , Cobalt/chemistry , Electrochemical Techniques/methods , Biosensing Techniques/methods , Sulfides/chemistry , Carbon Fiber/chemistry , Glucose/analysis , Glucose/chemistry , Nanopores , Oxidation-Reduction , Blood Glucose/analysis
3.
J Nanobiotechnology ; 22(1): 51, 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38321547

ABSTRACT

BACKGROUND: Allergic rhinitis (AR) is a prevalent immune-related allergic disease, and corticosteroid nasal sprays serve as the primary treatment for this patient population. However, their short duration of efficacy and frequent administration pose challenges, leading to drug wastage and potential adverse effects. To overcome these limitations, we devised a novel approach to formulate DEX-Gel by incorporating dexamethasone (DEX) into a blend of Pluronic F127, stearic acid (SA), and polyethylene glycol 400 (PEG400) to achieve sustained-release treatment for AR. RESULTS: Following endoscopic injection into the nasal mucosa of AR rats, DEX-Gel exhibited sustained release over a 14-day period. In vivo trials employing various assays, such as flow cytometry (FC), demonstrated that DEX-Gel not only effectively managed allergic symptoms but also significantly downregulated helper T-cells (TH) 2 and TH2-type inflammatory cytokines (e.g., interleukins 4, 5, and 13). Additionally, the TH1/TH2 cell ratio was increased. CONCLUSION: This innovative long-acting anti-inflammatory sustained-release therapy addresses the TH1/TH2 immune imbalance, offering a promising and valuable approach for the treatment of AR and other inflammatory nasal diseases.


Subject(s)
Rhinitis, Allergic , Th1 Cells , Humans , Rats , Animals , Mice , Delayed-Action Preparations/pharmacology , Th2 Cells , Rhinitis, Allergic/drug therapy , Cytokines , Anti-Inflammatory Agents/pharmacology , Disease Models, Animal , Ovalbumin , Mice, Inbred BALB C
4.
Small ; 19(4): e2204133, 2023 01.
Article in English | MEDLINE | ID: mdl-36420659

ABSTRACT

The acquired resistance to Osimertinib (AZD9291) greatly limits the clinical benefit of patients with non-small cell lung cancer (NSCLC), whereas AZD9291-resistant NSCLCs are prone to metastasis. It's challenging to overcome AZD9291 resistance and suppress metastasis of NSCLC simultaneously. Here, a nanocatalytic sensitizer (VF/S/A@CaP) is proposed to deliver Vitamin c (Vc)-Fe(II), si-OTUB2, ASO-MALAT1, resulting in efficient inhibition of tumor growth and metastasis of NSCLC by synergizing with AHP-DRI-12, an anti-hematogenous metastasis inhibitor by blocking the amyloid precursor protein (APP)/death receptor 6 (DR6) interaction designed by our lab. Fe2+ released from Vc-Fe(II) generates cytotoxic hydroxyl radicals (•OH) through Fenton reaction. Subsequently, glutathione peroxidase 4 (GPX4) is consumed to sensitize AZD9291-resistant NSCLCs with high mesenchymal state to ferroptosis due to the glutathione (GSH) depletion caused by Vc/dehydroascorbic acid (DHA) conversion. By screening NSCLC patients' samples, metastasis-related targets (OTUB2, LncRNA MALAT1) are confirmed. Accordingly, the dual-target knockdown plus AHP-DRI-12 significantly suppresses the metastasis of AZD9291-resistant NSCLC. Such modality leads to 91.39% tumor inhibition rate in patient-derived xenograft (PDX) models. Collectively, this study highlights the vulnerability to ferroptosis of AZD9291-resistant tumors and confirms the potential of this nanocatalytic-medicine-based modality to overcome critical AZD9291 resistance and inhibit metastasis of NSCLC simultaneously.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Ferroptosis , Lung Neoplasms , RNA, Long Noncoding , Humans , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , ErbB Receptors/metabolism , Drug Resistance, Neoplasm/genetics , Ferrous Compounds , Cell Line, Tumor
5.
J Nanobiotechnology ; 21(1): 17, 2023 Jan 16.
Article in English | MEDLINE | ID: mdl-36647107

ABSTRACT

BACKGROUND: Superhydrophobic substrate modifications are an effective way to improve SERS sensitivity by concentrating analyte molecules into a small surface area. However, it is difficult to manipulate low-volume liquid droplets on superhydrophobic substrates. RESULTS: To overcome this limitation, we deposited a hydrophilic Ti3C2Tx film on a superhydrophobic ZnO nanorod array to create a SERS substrate with improved analyte affinity. Combined with its interfacial charge transfer properties, this enabled a rhodamine 6G detection limit of 10-11 M to be achieved. In addition, the new SERS substrate showed potential for detection of biological macromolecules, such as microRNA. CONCLUSION: Combined with its facile preparation, the SERS activity of ZnO/Ti3C2Tx suggests it may provide an ultrasensitive environmental pollutant-monitoring and effective substrate for biological analyte detection.


Subject(s)
Environmental Pollutants , Zinc Oxide , Zinc Oxide/chemistry , Spectrum Analysis, Raman , Titanium/chemistry , Silver/chemistry , Hydrophobic and Hydrophilic Interactions , Environmental Pollutants/analysis
6.
J Nanobiotechnology ; 20(1): 50, 2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35078498

ABSTRACT

BACKGROUND: Although cisplatin-based chemotherapy has been used as the first-line treatment for ovarian cancer (OC), tumor cells develop resistance to cisplatin during treatment, causing poor prognosis in OC patients. Studies have demonstrated that overactivation of the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway is involved in tumor chemoresistance and that overexpression of microRNA-497 (miR497) may overcome OC chemotherapy resistance by inhibiting the mTOR pathway. However, the low transcriptional efficiency and unstable chemical properties of miR497 limit its clinical application. Additionally, triptolide (TP) was confirmed to possess a superior killing effect on cisplatin-resistant cell lines, partially through inhibiting the mTOR pathway. Even so, the clinical applications of TP are restricted by serious systemic toxicity and weak water solubility. RESULTS: Herein, whether the combined application of miR497 and TP could further overcome OC chemoresistance by synergically suppressing the mTOR signaling pathway was investigated. Bioinspired hybrid nanoparticles formed by the fusion of CD47-expressing tumor exosomes and cRGD-modified liposomes (miR497/TP-HENPs) were prepared to codeliver miR497 and TP. In vitro results indicated that the nanoparticles were efficiently taken up by tumor cells, thus significantly enhancing tumor cell apoptosis. Similarly, the hybrid nanoparticles were effectively enriched in the tumor areas and exerted significant anticancer activity without any negative effects in vivo. Mechanistically, they promoted dephosphorylation of the overactivated PI3K/AKT/mTOR signaling pathway, boosted reactive oxygen species (ROS) generation and upregulated the polarization of macrophages from M2 to M1 macrophages. CONCLUSION: Overall, our findings may provide a translational strategy to overcome cisplatin-resistant OC and offer a potential solution for the treatment of other cisplatin-resistant tumors.


Subject(s)
Exosomes , MicroRNAs , Nanoparticles , Ovarian Neoplasms , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cisplatin/pharmacology , Cisplatin/therapeutic use , Diterpenes , Drug Resistance, Neoplasm , Epoxy Compounds , Exosomes/metabolism , Humans , Liposomes/pharmacology , MicroRNAs/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Phenanthrenes , Phosphatidylinositol 3-Kinases/metabolism
7.
J Nanobiotechnology ; 19(1): 55, 2021 Feb 25.
Article in English | MEDLINE | ID: mdl-33632232

ABSTRACT

BACKGROUND: Triple negative breast cancer (TNBC) is one of the most biologically aggressive breast cancers and lacks effective treatment options, resulting in a poor prognosis. Therefore, studies aiming to explore new therapeutic strategies for advanced TNBC are urgently needed. According to recent studies, microRNA-124 (miR124) not only inhibits tumour growth but also increases the sensitivity of TNBC to paclitaxel (PTX), suggesting that a platform combining PTX and miR124 may be an advanced solution for TNBC. RESULTS: Herein, we constructed a stepped cleavable calcium phosphate composite lipid nanosystem (CaP/LNS) to codeliver PTX and miR124 (PTX/miR124-NP). PTX/miR124-NP exhibited superior tumor microenvironment responsive ability, in which the surface PEG layer was shed in the mildly acidic environment of tumor tissues and exposed oligomeric hyaluronic acid (o-HA) facilitated the cellular uptake of CaP/LNS by targeting the CD44 receptor on the surface of tumor cells. Inside tumour cells, o-HA detached from CaP/LNS due to the reduction of disulfide bonds by glutathione (GSH) and inhibited tumour metastasis. Then, PTX and miR124 were sequentially released from CaP/LNS and exerted synergistic antitumour effects by reversing the Epithelial-Mesenchymal Transition (EMT) process in MDA-MB-231 cells. Moreover, PTX/miR124-NP showed significant antitumour efficiency and excellent safety in mice bearing MDA-MB-231 tumours. CONCLUSION: Based on these results, the codelivery of PTX and miR124 by the CaP/LNS nanosystem might be a promising therapeutic strategy for TNBC.


Subject(s)
MicroRNAs/pharmacology , Paclitaxel/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement , Female , Hyaluronan Receptors , Hyaluronic Acid/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Paclitaxel/chemistry , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment/drug effects
8.
J Nanobiotechnology ; 19(1): 165, 2021 May 31.
Article in English | MEDLINE | ID: mdl-34059068

ABSTRACT

BACKGROUND: Overexpressed vascular endothelial growth factor A (VEGFA) and phosphorylated signal transducer and activator of transcription 3 (P-STAT3) cause unrestricted tumor growth and angiogenesis of breast cancer (BRCA), especially triple-negative breast cancer (TNBC). Hence, novel treatment strategy is urgently needed. RESULTS: We found sphingosine 1 phosphate receptor 1 (S1PR1) can regulate P-STAT3/VEGFA. Database showed S1PR1 is highly expressed in BRCA and causes the poor prognosis of patients. Interrupting the expression of S1PR1 could inhibit the growth of human breast cancer cells (MCF-7 and MDA-MB-231) and suppress the angiogenesis of human umbilical vein endothelial cells (HUVECs) via affecting S1PR1/P-STAT3/VEGFA axis. Siponimod (BAF312) is a selective antagonist of S1PR1, which inhibits tumor growth and angiogenesis in vitro by downregulating the S1PR1/P-STAT3/VEGFA axis. We prepared pH-sensitive and tumor-targeted shell-core structure nanoparticles, in which hydrophilic PEG2000 modified with the cyclic Arg-Gly-Asp (cRGD) formed the shell, hydrophobic DSPE formed the core, and CaP (calcium and phosphate ions) was adsorbed onto the shell; the nanoparticles were used to deliver BAF312 (BAF312@cRGD-CaP-NPs). The size and potential of the nanoparticles were 109.9 ± 1.002 nm and - 10.6 ± 0.056 mV. The incorporation efficacy for BAF312 was 81.4%. Results confirmed BAF312@cRGD-CaP-NP could dramatically inhibit tumor growth and angiogenesis in vitro and in MDA-MB-231 tumor-bearing mice via downregulating the S1PR1/P-STAT3/VEGFA axis. CONCLUSIONS: Our data suggest a potent role for BAF312@cRGD-CaP-NPs in treating BRCA, especially TNBC by downregulating the S1PR1/P-STAT3/VEGFA axis.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Azetidines/pharmacology , Benzyl Compounds/pharmacology , Nanoparticles/chemistry , STAT3 Transcription Factor/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Triple Negative Breast Neoplasms/blood supply , Triple Negative Breast Neoplasms/drug therapy , Vascular Endothelial Growth Factor A/metabolism , Animals , Azetidines/chemistry , Benzyl Compounds/chemistry , Cell Line, Tumor , Down-Regulation/drug effects , Endothelial Cells/metabolism , Humans , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , STAT3 Transcription Factor/genetics , Sphingosine-1-Phosphate Receptors/genetics , Triple Negative Breast Neoplasms/genetics , Vascular Endothelial Growth Factor A/genetics
9.
J Nanobiotechnology ; 17(1): 125, 2019 Dec 23.
Article in English | MEDLINE | ID: mdl-31870362

ABSTRACT

BACKGROUND: Multidrug resistance (MDR) is a pressing obstacle in clinical chemotherapy for breast cancer. Based on the fact that the drug efflux is an important factor in MDR, we designed a codelivery system to guide the drug efflux inhibitor verapamil (VRP) and the chemotherapeutic agent novantrone (NVT) synergistically into breast cancer cells to reverse MDR. RESULTS: This co-delivery system consists of following components: the active targeting peptide RGD, an inorganic calcium phosphate (CaP) shell and an organic inner core. VRP and NVT were loaded into CaP shell and phosphatidylserine polyethylene glycol (PS-PEG) core of nanoparticles (NPs) separately to obtain NVT- and VRP-loaded NPs (NV@CaP-RGD). These codelivered NPs allowed VRP to prevent the efflux of NVT from breast cancer cells by competitively combining with drug efflux pumps. Additionally, NV@CaP-RGD was effectively internalized into breast cancer cells by precise delivery through the effects of the active targeting peptides RGD and EPR. The pH-triggered profile of CaP was also able to assist the NPs to successfully escape from lysosomes, leading to a greatly increased effective intracellular drug concentration. CONCLUSION: The concurrent administration of VRP and NVT by organic/inorganic NPs is a promising therapeutic approach to reverse MDR in breast cancer.


Subject(s)
Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Mitoxantrone/chemistry , Nanocapsules/chemistry , Verapamil/chemistry , Animals , Calcium Phosphates/chemistry , Cell Line, Tumor , Cell Membrane Permeability , Cell Survival , Drug Compounding/methods , Drug Liberation , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Drug Therapy, Combination/methods , Female , Humans , Mice, Inbred BALB C , Mice, Nude , Mitoxantrone/pharmacology , Molecular Targeted Therapy , Oligopeptides/chemistry , Oligopeptides/metabolism , Phosphatidylserines/chemistry , Polyethylene Glycols/chemistry , Verapamil/metabolism
10.
Nanomedicine ; 21: 102062, 2019 10.
Article in English | MEDLINE | ID: mdl-31344501

ABSTRACT

Primary hepatocellular carcinoma (HCC) is a common malignant tumor. Surgery is the main treatment, but HCC patients have a potential risk of tumor recurrence. Besides, many limitations arise during the application of single first-line antitumor drugs. Here, we selected Pluronic F-127 and sodium alginate (SA) to prepare a thermosensitive gel (Gel). The optimal synergistic ratio of PTX and DOX on the SMMC-7721 cells was 1: 2 (w/w), calculated by the Chou-Talalay analysis. Then, PTX and DOX coloaded liposomes (PD-LPs) with such drugs ratios presented enhanced anticancer ability in vitro. Upon local injection, the PD-LPs Gel formed a nanoparticles reservoir at tumor via sol-gel transformation, while exhibiting a long-term effective anti-tumor ability in vivo. The relative tumor volume after the PD-LPs Gel treatment was reduced over 62%. Effective mitochondria related apoptosis induction was observed. Therefore, the local delivery of PD-LPs Gel can be a promising alternative method for the HCC therapy.


Subject(s)
Apoptosis/drug effects , Carcinoma, Hepatocellular , Doxorubicin , Liver Neoplasms, Experimental , Mitochondria, Liver , Nanocomposites , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Humans , Liposomes , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Mice , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Nanocomposites/chemistry , Nanocomposites/therapeutic use , Xenograft Model Antitumor Assays
11.
Int J Mol Sci ; 18(4)2017 Apr 13.
Article in English | MEDLINE | ID: mdl-28406431

ABSTRACT

In general, atherosclerosis is considered to be a form of chronic inflammation. Dexamethasone has anti-inflammatory effects in atherosclerosis, but it was not considered for long-term administration on account of a poor pharmacokinetic profile and adverse side effects. Nanoparticles in which drugs can be dissolved, encapsulated, entrapped or chemically attached to the particle surface have abilities to incorporate dexamethasone and to be used as controlled or targeted drug delivery system. Long circulatory polymeric nanoparticles present as an assisting approach for controlled and targeted release of the encapsulated drug at the atherosclerotic site. Polymeric nanoparticles combined with ultrasound (US) are widely applied in cancer treatment due to their time applications, low cost, simplicity, and safety. However, there are few studies on atherosclerosis treatment using polymeric nanoparticles combined with US. In this study, targeted dexamethasone acetate (DA)-loaded poly (lactide-glycolide)-polyethylene glycol-cRGD (PLGA-PEG-cRGD) nanoparticles (DA-PLGA-PEG-cRGD NPs) were prepared by the emulsion-evaporation method using cRGD modified PLGA-PEG polymeric materials (PLGA-PEG-cRGD) prepared as the carrier. The average particle size of DA-PLGA-PEG-cRGD NPs was 221.6 ± 0.9 nm. Morphology of the nanoparticles was spherical and uniformly dispersed. In addition, the DA released profiles suggested that ultrasound could promote drug release from the nanocarriers and accelerate the rate of release. In vitro, the cellular uptake process of fluorescein isothiocyanate (FITC)@DA-PLGA-PEG-cRGD NPs combined with US into the damaged human umbilical vein endothelial cells (HUVECs) indicated that US promoted rapid intracellular uptake of FITC@DA- PLGA-PEG-cRGD NPs. The cell viability of DA-PLGA-PEG-cRGD NPs combined with US reached 91.9% ± 0.2%, which demonstrated that DA-PLGA-PEG-cRGD NPs combined with US had a positive therapeutic effect on damaged HUVECs. Overall, DA-PLGA-PEG-cRGD NPs in combination with US may provide a promising drug delivery system to enhance the therapeutic effects of these chemotherapeutics at the cellular level.


Subject(s)
Dexamethasone/analogs & derivatives , Drug Carriers/chemistry , Lactic Acid/chemistry , Nanoparticles/chemistry , Oligopeptides/chemistry , Polyethylene Glycols/chemistry , Polyglycolic Acid/chemistry , Cell Survival/drug effects , Dexamethasone/chemistry , Dexamethasone/pharmacology , Drug Liberation , Fluorescein-5-isothiocyanate/chemistry , Human Umbilical Vein Endothelial Cells , Humans , Lipoproteins, LDL/toxicity , Magnetic Resonance Spectroscopy , Microscopy, Confocal , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer , Sonication
12.
Phys Chem Chem Phys ; 18(17): 11986-99, 2016 04 28.
Article in English | MEDLINE | ID: mdl-27067001

ABSTRACT

In a previous study, a novel biodegradable multiblock copolymer, monomethoxy(poly-ethylene glycol)-poly(d,l-lactide-co-glycolide)-poly(l-lysine) (PEAL), was developed as a new drug carrier material. It is imperative to study the biocompatibility and degradation behavior of PEAL to pave the way for clinical applications. Here, we systematically demonstrated that the PEAL copolymer has the appropriate hydrophilicity and biosafety. The degradation rate of the PEAL films was obtained by observing changes in mass, molecular weight (Mw), Mw distribution and degradation products. The degradation rate was observed to have a highly positive correlation with the pH of the medium and negative correlation with the ratio of lactic acid to glycolic acid (LA/GA). Cytotoxicity tests indicated that the degradation products of the copolymer were non-toxic to cells. In zebrafish embryos, the PEAL nanoparticles had no obvious impact on heart rate, production of reactive oxygen species, mortality, or cell apoptosis, and they were observed to have a long circulation time. Therefore, the PEAL copolymer has great potential for use as a drug carrier material.


Subject(s)
Drug Carriers/metabolism , Polyesters/metabolism , Polyethylene Glycols/metabolism , Polylysine/metabolism , Animals , Blood Coagulation/drug effects , Blood Platelets/cytology , Blood Platelets/drug effects , Cell Line , Complement Activation/drug effects , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/toxicity , Heart Rate/drug effects , Humans , Hydrophobic and Hydrophilic Interactions , Nanoparticles/analysis , Nanoparticles/chemistry , Nanoparticles/metabolism , Nanoparticles/toxicity , Polyesters/chemistry , Polyesters/pharmacokinetics , Polyesters/toxicity , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/toxicity , Polylysine/chemistry , Polylysine/pharmacokinetics , Polylysine/toxicity , Reactive Oxygen Species/metabolism , Zebrafish
13.
J Nanosci Nanotechnol ; 15(12): 9777-87, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26682412

ABSTRACT

In this work, we have reported the preparation and optimization of paclitaxel (PTX) and temozolomide (TMZ) loaded monomethoxy (polyethylene glycol)-poly(D, L-lactide-co-glycolide) (mPEG-PLGA) nanocomposite which is a thermo-sensitive gel delivery system to glioblastoma. We utilized the orthogonal design and homogeneous design for the optimal drug-loaded nanoparticles (NPs) and composite gel prescription, respectively. The physicochemical characteristics of NPs and rheological properties of the gel were analyzed. Then the in vitro release of the gel was determined with a membrane-less diffusion system. Finally, the cytotoxic and apoptosis-inducing effects of the gel on the human malignant glioblastoma cell line U87 and C6 rat glioblastoma cell line were evaluated by MTT and flow cytometry apoptosis assay, respectively. The transmission electron microscopy (TEM) analysis revealed the optimized NPs with a relatively uniform diameter and distribution. The homogeneous design and rheological determination showed that the optimized gel prescription was 250 mg/mL Pluronic F127 (F127), 0.5% hydroxy propyl methylcellulose (HPMC-100M), 0.5% Pluronic F68 (F68), 0.5% sodium alginate (SA) and suitable NPs, which possessed the appropriate gelation behaviors: gelation temperature 28.01 degrees C, gelation time 127.1 s and corrosion speed 0.1892 g/cm2 x hr; and rheological properties: suitable elasticity modulus, viscosity modulus and low phase angle. The in vitro results suggested that the PTX and TMZ were sustainedly released from nanoparticles or the composite gel, and the release and elimination time greatly prolonged; and the composite gel possessed much higher growth-inhibiting effect and apoptosis-inducing rate in U87 and C6 cells than other formulations. These findings demonstrated that the optimal gel was a promising delivery system for the interstitial chemotherapy to glioblastoma.


Subject(s)
Dacarbazine/analogs & derivatives , Drug Carriers/chemistry , Glioblastoma/drug therapy , Nanocomposites/chemistry , Paclitaxel/pharmacology , Animals , Cell Line, Tumor , Dacarbazine/pharmacology , Humans , Polyesters/chemistry , Polyethylene Glycols/chemistry , Rats , Temozolomide
14.
Int J Mol Sci ; 16(5): 9573-87, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25927579

ABSTRACT

The prostate specific membrane antigen (PSMA) is broadly overexpressed on prostate cancer (PCa) cell surfaces. In this study, we report the synthesis, characterization, in vitro binding assay, and in vivo magnetic resonance imaging (MRI) evaluation of PSMA targeting superparamagnetic iron oxide nanoparticles (SPIONs). PSMA-targeting polypeptide CQKHHNYLC was conjugated to SPIONs to form PSMA-targeting molecular MRI contrast agents. In vitro studies demonstrated specific uptake of polypeptide-SPIONs by PSMA expressing cells. In vivo MRI studies found that MRI signals in PSMA-expressing tumors could be specifically enhanced with polypeptide-SPION, and further Prussian blue staining showed heterogeneous deposition of SPIONs in the tumor tissues. Taken altogether, we have developed PSMA-targeting polypeptide-SPIONs that could specifically enhance MRI signal in tumor-bearing mice, which might provide a new strategy for the molecular imaging of PCa.


Subject(s)
Antigens, Surface/chemistry , Ferric Compounds/chemistry , Glutamate Carboxypeptidase II/chemistry , Metal Nanoparticles/chemistry , Prostatic Neoplasms/diagnosis , Animals , Cell Line, Tumor , Contrast Media/chemistry , Ferrocyanides/chemistry , Humans , Lactic Acid/chemistry , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Nanotechnology , Neoplasm Transplantation , Peptides/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Prostatic Neoplasms/pathology , Protein Binding , Signal Transduction
15.
J Nanosci Nanotechnol ; 14(6): 4078-81, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24738354

ABSTRACT

Colloidal gold is extensively used for molecular sensing because that the surface plasmon resonance (SPR) bands are affected by changes in the dielectric properties in the close vicinity of these structures due to the binding of ligands to the corresponding receptor molecules immobilized onto the nanostructures through chemi- or physisorption. We describe a simple method for the detection of Alpha-Fetoprotein-L3 which is a new generation of tumor marker for hepatocellular carcinoma (HCC) based on the aggregation of Lens culinaris agglutinin (LCA) Immobilized Gold Nanoparticles. The LCA conjugated GNPs obtained were 15-20 nm in diameter. The visible color change of the gold nanoparticles from purple to blue on interaction with 100 ng/mL of AFP-L3 is the principle applied here for the sensing of AFP-L3 level. UV/Vis spectroscopy also allows assay monitoring by quantifying the red shift of the plasmon resonance wavelength. With this method, the protein AFP-L3 can be rapidly detected as demanded for clinical diagnosis.


Subject(s)
Gold/chemistry , Metal Nanoparticles/chemistry , Microscopy, Electron/methods , Nanoconjugates/chemistry , Plant Lectins/chemistry , Spectrophotometry, Ultraviolet/methods , alpha-Fetoproteins/analysis , Adsorption , Biomarkers, Tumor/analysis , Gold/analysis , Metal Nanoparticles/analysis , Protein Binding , alpha-Fetoproteins/chemistry
16.
J Nanosci Nanotechnol ; 14(7): 4843-50, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24757952

ABSTRACT

Pluronic-poly[alpha-(4-aminobutyl)-1-glycolic acid] (Pluronic-PAGA) with different types of Pluronic, the different molecule weight of PAGA, and the different molar ratios of Pluronic to PAGA were synthesized. These materials were bio-degradable, amphiphilic, could be degraded into non-toxic small molecules and could be used to carry drugs. 5-Fluorouracil (5-Fu) loaded Pluronic-PAGA micelle-like nanoparticles (5-Fu loaded P-PAGA NPs) were prepared by a simple self-assembly method, and characterized by dynamic light scattering, transmission electron microscope. The degradation and release characteristics have also been studied in this paper. With the time passing, the 5-Fu loaded P-PAGA NPs degraded into smaller ones with the similar characteristics of the original NPs. Both the types of Pluronic and the molecule weight of the PAGA affected the releasing progresses. It was found that 5-Fu loaded P-PAGA NPs exhibited high growth inhibitory effect on human gastric cancer cells by MTT assay. The cellular uptake of Rhodamine B loaded P-PAGA NPs was higher than free Rhodamine B. This study suggested that the Pluronic-PAGA with acceptable drug entrapment efficiencies, drug loading efficiencies and tunable release profiles could offer an alternative carrier for 5-Fu delivery and have the potential for the delivery of other anti-tumour drug.


Subject(s)
Fluorouracil/administration & dosage , Fluorouracil/chemistry , Nanocapsules/administration & dosage , Nanocapsules/chemistry , Poloxamer/chemistry , Polyglycolic Acid/analogs & derivatives , Stomach Neoplasms/drug therapy , Antimetabolites, Antineoplastic/administration & dosage , Cell Line, Tumor , Diffusion , Humans , Micelles , Molecular Weight , Nanocapsules/ultrastructure , Particle Size , Polyglycolic Acid/chemistry , Stomach Neoplasms/pathology , Treatment Outcome
17.
J Mater Chem B ; 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38919121

ABSTRACT

A solvothermal method and the subsequent heat treatment process were developed to fabricate hollow ZnO particles with hierarchical pores on a large scale. The as-obtained hollow, porous ZnO microspheres with tunable sizes, high specific surface areas, pH sensitivity, antibacterial properties, and high adsorption capacities showed significant advantages for drug delivery. Sprayable hydrogels containing hollow, porous ZnO microspheres and curcumin nanoparticles (CNPs) were prepared to accelerate wound healing. The water-dispersed CNPs promoted both the migration of fibroblasts and angiogenesis and an aqueous solution of Pluronic F127 (a temperature-sensitive phase-change hydrogel material) was shown to be an effective choice for medical dressings. The experimental data suggest that hollow, porous ZnO microspheres can be loaded with additional CNPs to achieve continuous long-term therapeutic effects.

18.
J Control Release ; 371: 111-125, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38782064

ABSTRACT

In esophageal cancer (EC), clinical specimen testing has uncovered a significant increase in BTB and CNC homolog 1 (BACH1) expression and a shift towards an immunosuppressive environment, alongside a notable decrease in p53 protein expression. Therefore, therapeutic strategies focusing on BACH1 inhibition and p53 upregulation appear promising. Traditional oral treatments for EC lack precision and efficacy. Here, we propose a novel approach employing tumor-targeted nanoparticles (NPs) for drug delivery. However, the formation of a drug reservoir at the esophageal site, crucial for the sustained release of therapeutics, presents significant challenges in nano-delivery systems for EC treatment. To address this, we developed a thermosensitive hydrogel composed of F127 and tannic acid, serving as a vehicle for NP loading. These NPs, synthesized through the emulsion/volatization methods of mPEG-PLGA-PLL-cRGD, facilitate in situ drug delivery. Upon contacting esophageal tissue, the hydrogel transitions to a gel, adhering to the lining and enabling sustained release of encapsulated therapeutics. The formulation encompasses NPs laden with small interfering RNA targeting BACH1 (siBACH1) and the p53 activator PRIMA-1, creating a cohesive gel-nano system. Preliminary biological assessments demonstrate that this injectable, thermosensitive gel-nano system adheres effectively to esophageal tissue and targets EC cells. For better modeling clinical outcomes, a patient-derived organoid xenograft (PDOX) model was innovated, involving transplantation of EC-derived organoids into humanized mice, reconstructed with peripheral blood mononuclear cells (PBMCs). Post-treatment analysis showed substantial EC growth inhibition (89.51% tumor inhibition rate), significant BACH1 level reduction, restored anti-tumor immune responses, and pronounced tumor apoptosis. In summary, our study introduces a thermosensitive gel-nano system for EC treatment via restoring p53 activity and boosting T-cell immunity, with potential for clinical application.


Subject(s)
Esophageal Neoplasms , Nanoparticles , Tumor Suppressor Protein p53 , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/immunology , Animals , Humans , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Cell Line, Tumor , Hydrogels/administration & dosage , Hydrogels/chemistry , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Female , Mice , Temperature , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Drug Delivery Systems
19.
Diagnostics (Basel) ; 14(7)2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38611585

ABSTRACT

BACKGROUND AND OBJECTIVE: The dynamic assessment of disease activity during the follow-up of patients with Crohn's disease (CD) remains a significant challenge. In this study, we aimed to identify the role of dynamic contrast-enhanced ultrasound (DCE-US) in the evaluation of activity of CD. METHODS: In the retrospective study, patients diagnosed with CD in our hospital were included. All the diagnoses were confirmed by clinical symptoms and ileocolonoscopical results. All patients underwent intestinal ultrasound and contrast-enhanced ultrasound (CEUS) examinations within 1 week of the ileocolonoscopy examinations. Acuson Sequoia (Siemens Healthineers, Mountain View, CA, USA) and Resona R9 Elite (Mindray Medical Systems, China) with curved array and Line array transducers were used. The CEUS examination was performed with SonoVue (Bracco SpA, Milan, Italy). DCE-US analysis was performed by UltraOffice (version: 0.3-2010, Mindray Medical Systems, China) software. Two regions of interest (ROIs) were set in the anterior section of the infected bowel wall and its surrounding normal bowel wall 2 cm distant from the inflamed area. Time-intensity curves (TICs) were generated and quantitative perfusion parameters were obtained after curve fittings. The Simple Endoscopic Score for Crohn's disease (SES-CD) was regarded as the reference standard to evaluate the activity of CD. The receiver operating characteristic curve (ROC) analyses were used to determine the diagnostic efficiency of DCE-US quantitative parameters. RESULTS: From March 2023 to November 2023, 52 CD patients were included. According to SES-CD score, all patients were divided into active group with the SES-CD score > 5 (n = 39) and inactive group SES-CD score < 5 (n = 13). Most of the active CD patients showed bowel wall thickness (BWT) > 4.2 mm (97.4%, 38/39) or mesenteric fat hypertrophy (MFH) on intestinal ultrasound (US) scan (69.2%, 27/39). Color Doppler signal of the bowel wall mostly showed spotty or short striped blood flow signal in active CD patients (56.4%, 22/39). According to CEUS enhancement patterns, most active CD patients showed a complete hyperenhancement of the entire intestinal wall (61.5%, 24/39). The TICs of active CD showed an earlier enhancement, higher peak intensity, and faster decline. Among all CEUS quantitative parameters, amplitude-derived parameters peak enhancement (PE), wash-in area under the curve (WiAUC), wash-in rate (WiR), wash-in perfusion index (WiPI), and wash-out rate (WoR) were significantly higher in active CD than in inactive CD (p < 0.05). The combined AUROC of intestinal ultrasound features and DCE-US quantitative perfusion parameters in the diagnosis of active CD was 0.987, with 97.4% sensitivity, 100% specificity, and 98.1% accuracy. CONCLUSIONS: DCE-US with quantitative perfusion parameters is a potential useful noninvasive imaging method to evaluate the activity of Crohn's disease.

20.
Adv Sci (Weinh) ; 11(20): e2308310, 2024 May.
Article in English | MEDLINE | ID: mdl-38520730

ABSTRACT

CD47 blockade has emerged as a promising immunotherapy against liver cancer. However, the optimization of its antitumor effectiveness using efficient drug delivery systems or combinations of therapeutic agents remains largely incomplete. Here, patients with liver cancer co-expressing CD47 and CDC7 (cell division cycle 7, a negative senescence-related gene) are found to have the worst prognosis. Moreover, CD47 is highly expressed, and senescence is inhibited after the development of chemoresistance, suggesting that combination therapy targeting CD47 and CDC7 to inhibit CD47 and induce senescence may be a promising strategy for liver cancer. The efficacy of intravenously administered CDC7 and CD47 inhibitors is limited by low uptake and short circulation times. Here, inhibitors are coloaded into a dual-targeted nanosystem. The sequential release of the inhibitors from the nanosystem under acidic conditions first induces cellular senescence and then promotes immune responses. In an in situ liver cancer mouse model and a chemotherapy-resistant mouse model, the nanosystem effectively inhibited tumor growth by 90.33% and 85.15%, respectively. Overall, the nanosystem in this work achieved the sequential release of CDC7 and CD47 inhibitors in situ to trigger senescence and induce immunotherapy, effectively combating liver cancer and overcoming chemoresistance.


Subject(s)
CD47 Antigen , Liver Neoplasms , Animals , Mice , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , CD47 Antigen/metabolism , Humans , Disease Models, Animal , Cellular Senescence/drug effects , Cell Cycle Proteins/metabolism , Immunotherapy/methods , Drug Delivery Systems/methods , Nanoparticles , Immunologic Factors/pharmacology , Cell Line, Tumor , Immunomodulating Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL