Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
J Cell Sci ; 134(8)2021 04 15.
Article in English | MEDLINE | ID: mdl-33758077

ABSTRACT

Autophagy is considered to be an important switch for facilitating normal to malignant cell transformation during colorectal cancer development. Consistent with other reports, we found that the membrane receptor Neuropilin1 (NRP1) is greatly upregulated in colon cancer cells that underwent autophagy upon glucose deprivation. However, the mechanism underlying NRP1 regulation of autophagy is unknown. We found that knockdown of NRP1 inhibits autophagy and largely upregulates the expression of aldo-keto reductase family 1 B10 (AKR1B10). Moreover, we demonstrated that AKR1B10 interacts with and inhibits the nuclear importation of glyceraldehyde-3-phosphate dehydrogenase (GAPDH), and then subsequently represses autophagy. Interestingly, we also found that an NADPH-dependent reduction reaction could be induced when AKR1B10 interacts with GAPDH, and the reductase activity of AKR1B10 is important for its repression of autophagy. Together, our findings unravel a novel mechanism of NRP1 in regulating autophagy through AKR1B10.


Subject(s)
Aldehyde Reductase , Colonic Neoplasms , Aldehyde Reductase/genetics , Aldo-Keto Reductases , Autophagy , Colonic Neoplasms/genetics , Glucose , Glyceraldehyde-3-Phosphate Dehydrogenases , Humans
2.
Mol Pharm ; 19(7): 2607-2619, 2022 07 04.
Article in English | MEDLINE | ID: mdl-35485954

ABSTRACT

Chlorin e6-C-15-ethyl ester (LS-HB), a newly identified photosensitizer, was isolated from chlorin e6. The mechanism of tumor cell death induced by photodynamic therapy with LS-HB (LS-HB-PDT) is still unknown. Here, we investigated the photophysical properties of LS-HB, evaluated the antitumor effect on melanoma in vitro and in vivo, and explored its possible mechanisms. LS-HB not only has an optimal spectral band of red wavelength (660 nm) for photosensitization but also has favorable photostability. More importantly, LS-HB-PDT elicited a potent dose-dependent phototoxic effect in vitro. We discovered that LS-HB located in the mitochondria of B16F10 cells was able to generate excess reactive oxygen species, which subsequently resulted in mitochondrial membrane potential loss and induced apoptosis via caspase-9 and caspase-3 pathways. Moreover, PDT with LS-HB markedly inhibited the growth of melanoma in vivo. Therefore, LS-HB is expected to be an effective potential photosensitizer in antitumor therapy.


Subject(s)
Melanoma , Photochemotherapy , Porphyrins , Apoptosis , Cell Line, Tumor , Cell Proliferation , Humans , Melanoma/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Porphyrins/pharmacology , Porphyrins/therapeutic use , Reactive Oxygen Species/metabolism
3.
Mol Pharm ; 19(5): 1356-1367, 2022 05 02.
Article in English | MEDLINE | ID: mdl-35420039

ABSTRACT

Selectively inducing tumor thrombosis and subsequent necrosis is a novel and promising antitumor strategy. We have previously designed a targeting procoagulant protein, called tTF-EG3287, which is a fusion of a truncated tissue factor (tTF) with EG3287, a short peptide against the neuropilin-1 (NRP1) binding site of vascular endothelial growth factor-A 165 (VEGF-A 165). However, off-target effects and high-dose requirements limit the further use of tTF-EG3287 in antitumor therapy. Therefore, we encapsulated tTF-EG3287 into poly(2-ethyl-2-oxazoline)-distearoyl phosphatidyl ethanolamine (PEOz-DSPE)-modified liposomes to construct pH-responsive liposomes as a novel vascular embolization agent, called tTF-EG3287@Liposomes. The liposomes had an average particle size of about 100 nm and showed considerable drug-loading capacity, encapsulation efficiency, and biocompatibility. Under the stimulation of acidic microenvironments (pH 6.5), the lipid membrane of tTF-EG3287@Liposomes collapsed, and the cumulative drug release rate within 72 h was 83 ± 1.26%. When administered to a mouse model of hepatocellular carcinoma (HCC), tTF-EG3287@Liposomes showed prolonged retention and enhanced accumulation in the tumor as well as a superior antitumor effec, compared with tTF-EG3287. This study demonstrates the potential of tTF-EG3287@Liposomes as a novel embolic agent for solid tumors and provides a new strategy for tumor-targeted infarction therapy.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Hydrogen-Ion Concentration , Liposomes/chemistry , Liver Neoplasms/drug therapy , Mice , Thromboplastin , Tumor Microenvironment , Vascular Endothelial Growth Factor A
4.
Electrophoresis ; 41(15): 1273-1279, 2020 08.
Article in English | MEDLINE | ID: mdl-32358896

ABSTRACT

Mutations in the potassium channel genes may be linked to the development of epilepsy and affect the blood potassium levels. Therefore, accurate determination of potassium in the blood will be critical to diagnose the cause of epilepsy. CE is a competent technique for the fast detection of multiple ions, but complicated matrices of a blood sample may cause significant variation of migration times and the peak shape. In this work, a procedure for rapid stabilization of the capillary inner surface through preflushing of a blood sample was employed. The process takes only 40 min for a capillary and then it can be used for more than 2 weeks. No pretreatment of the blood sample or other surface modification of the capillary is needed for the analysis. The RSDs of the migration time and peak area were reduced to 1.5 and 5.1% from 12.6 and 14.5%, respectively. The proposed method has been successfully applied to the determination of the potassium contents in the blood sample of patients with epilepsy at different stages. The recoveries of potassium ions in these blood samples are in a range from 86.5 to 104.5%.


Subject(s)
Electrophoresis, Capillary/methods , Epilepsy/diagnosis , Potassium/blood , Blood Specimen Collection , Humans , Limit of Detection , Linear Models , Reproducibility of Results
5.
Mol Pharm ; 16(5): 1982-1998, 2019 05 06.
Article in English | MEDLINE | ID: mdl-30892898

ABSTRACT

Locating nanomedicines at the active sites plays a pivotal role in the nanoparticle-based cancer therapy field. Herein, a multifunctional nanotherapeutic is designed by using graphene oxide (GO) nanosheets with rich carboxyl groups as the supporter for hyaluronic acid (HA)-methotrexate (MTX) prodrug modification via an adipicdihydrazide cross-linker, achieving synergistic multistage tumor-targeting and combined chemo-photothermal therapy. As a tumor-targeting biomaterial, HA can increase affinity of the nanocarrier toward CD44 receptor for enhanced cellular uptake. MTX, a chemotherapeutic agent, can also serve as a tumor-targeting enhancer toward folate receptor based on its similar structure with folic acid. The prepared nanosystems possess a sheet shape with a dynamic size of approximately 200 nm and pH-responsive drug release. Unexpectedly, the physiological stability of HA-MTX prodrug-decorated GO nanosystems in PBS, serum, and even plasma is more excellent than that of HA-decorated GO nanosystems, while both of them exhibit an enhanced photothermal effect than GO nanosheets. More importantly, because of good blood compatibility as well as reduced undesired interactions with blood components, HA-MTX prodrug-decorated GO nanosystems exhibited remarkably superior accumulation at the tumor sites by passive and active targeting mechanisms, achieving highly effective synergistic chemo-photothermal therapeutic effect upon near-infrared laser irradiation, efficient ablation of tumors, and negligible systemic toxicity. Hence, the HA-MTX prodrug-decorated hybrid nanosystems have a promising potential for synergistic multistage tumor-targeting therapy.


Subject(s)
Drug Delivery Systems/methods , Graphite/chemistry , Nanoconjugates/chemistry , Photochemotherapy/methods , Uterine Cervical Neoplasms/therapy , Adipates/chemistry , Adipates/metabolism , Animals , Cell Survival/drug effects , Drug Liberation , Drug Stability , Drug Synergism , Female , Graphite/metabolism , HeLa Cells , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , MCF-7 Cells , Methotrexate/chemistry , Methotrexate/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , NIH 3T3 Cells , Prodrugs/chemistry , Prodrugs/metabolism , Tissue Distribution , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
6.
Anticancer Drugs ; 30(5): 441-450, 2019 06.
Article in English | MEDLINE | ID: mdl-30807552

ABSTRACT

In recent decades, selectively inducing tumor vascular thrombosis, followed by necrosis of tumor tissues has been a promising and potential anticancer strategy. In this report, we prepared a kind of vascular targeting drug that consists of anti-neuropilin-1 monoclonal antibody (anti-NRP-1 mAb) and truncated tissue factor (tTF). Anti-NRP-1 mAb could guide tTF to the surface of tumor vascular endothelial cells and lead to subsequent vascular embolization. This vascular targeting drug, which is also one of the antibody drug conjugates, was generated using a coupling method with water-soluble 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide and N-hydroxysulfosuccimide. Afterwards, in-vitro and in-vivo assays were performed to characterize its potential coagulation ability and antitumor activity. In-vitro experiments indicated that tTF-anti-NRP-1 monoclonal antibody (tTF-mAb) retained both the targeting activity of anti-NRP-1 mAb and the procoagulant activity of tTF. Live imaging system was used to assess its biodistribution and tumor-binding capability, which also yielded promising results. Furthermore, in-vivo studies showed that tTF-mAb was capable of significantly inducing tumor vascular thrombosis and inhibiting tumor growth in nude mice bearing subcutaneous xenografts, and histopathologic changes were rarely observed in normal organs.


Subject(s)
Antibodies, Monoclonal/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Neovascularization, Pathologic/prevention & control , Neuropilin-1/immunology , Thromboplastin/immunology , Thrombosis/prevention & control , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Apoptosis , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Humans , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Thrombosis/immunology , Thrombosis/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
J Clin Lab Anal ; 33(4): e22850, 2019 May.
Article in English | MEDLINE | ID: mdl-30758083

ABSTRACT

BACKGROUND: Neuropilins (Nrps) are a new type of broad-spectrum tumor marker. Currently, a method for accurate simultaneous quantification of Nrps is not available. We aimed to develop a bead-based and duplexed flow cytometric assay that could be used for accurate and simultaneous quantification of Nrp1 and Nrp2 for scientific research or clinical diagnosis. METHODS: We coupled anti-human Nrp1-11# mAb and anti-human Nrp2-C3 mAb to magnetic beads 18# and 25#, respectively. Capturing antibodies and detecting antibodies were then combined to detect Nrps by a bead-based Luminex assay, which was subsequently applied to quantify Nrps in clinical serum samples. RESULTS: The results showed that the detection value of Nrps ranged from 10 to 100 000 pg/mL for Nrp1 and from 25 to 100 000 pg/mL for Nrp2. The detection sensitivity reached 10 pg/mL for Nrp1 and 24.8 pg/mL for Nrp2. Intra-assay variances ranged from 1.0% to 2.6% for Nrp1 and from 2.9% to 4.0% for Nrp2, and interassay variances ranged from 1.5% to 6.4% for Nrp1 and from 4.2% to 8.1% for Nrp2. The Nrp1 and Nrp2 recoveries were 96.6%-103.6% and 95.6%-102.3%, respectively. Irrelevant antigens had no interference in the paired-detection system, and the mean fluorescence intensity (MFI) values were stable for months. CONCLUSION: A bead-based, duplexed flow cytometric assay (xMAP® technology) was developed to detect Nrp1 and Nrp2. The assay provided rapid, high-throughput results and was much more sensitive, specific, reproducible, and stable than existing assays. In addition, this assay could be applied in early-stage cancer screening, tumor malignancy analysis, and prognosis assessment.


Subject(s)
Immunoassay/methods , Neuropilin-1/blood , Neuropilin-2/blood , Antibodies, Monoclonal , Biomarkers, Tumor/blood , Biomarkers, Tumor/immunology , Biotinylation , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Humans , Immunoassay/instrumentation , Neoplasms/blood , Neuropilin-1/immunology , Neuropilin-2/immunology , Reproducibility of Results , Sensitivity and Specificity
8.
J Nanobiotechnology ; 16(1): 90, 2018 Nov 13.
Article in English | MEDLINE | ID: mdl-30424776

ABSTRACT

BACKGROUND: In the last decade, the biosynthesis of metal nanoparticles using organisms have received more and more considerations. However, the complex composition of organisms adds up to a great barrier for the characterization of biomolecules involved in the synthesis process and their biological mechanisms. RESULTS: In this research, we biosynthesized a kind of flower-shaped Au nanoclusters (Au NCs) using one definite component-epigallocatechin gallate (EGCG), which was the main biomolecules of green tea polyphenols. Possessing good stability for 6 weeks and a size of 50 nm, the Au NCs might be a successful candidate for drug delivery. Hence, both methotrexate (MTX) and doxorubicin (DOX) were conjugated to the Au NCs through a bridge of cysteine (Cys). The introduction of MTX provided good targeting property for the Au NCs, and the conjugation of DOX provided good synergistic effect. Then, a novel kind of dual-drug loaded, tumor-targeted and highly efficient drug delivery system (Au-Cys-MTX/DOX NCs) for combination therapy was successfully prepared. The TEM of HeLa cells incubated with Au-Cys-MTX/DOX NCs indicated that the Au-Cys-MTX/DOX NCs could indeed enter and kill cancer cells. The Au-Cys-MTX/DOX NCs also possessed good targeting effect to the FA-receptors-overpressed cancer cells both in vitro and in vivo. Importantly, the Au-Cys-MTX/DOX NCs resulted in an excellent anticancer activity in vivo with negligible side effects. CONCLUSIONS: These results suggest that the biosynthesized Au-Cys-MTX/DOX NCs could be a potential carrier with highly efficient anticancer properties for tumor-targeted drug delivery.


Subject(s)
Antineoplastic Agents/chemistry , Catechin/analogs & derivatives , Drug Carriers/chemistry , Gold/chemistry , Metal Nanoparticles/chemistry , Animals , Antineoplastic Agents/pharmacology , Catechin/chemistry , Cell Survival/drug effects , Delayed-Action Preparations , Doxorubicin/chemistry , Doxorubicin/pharmacology , Drug Liberation , HeLa Cells , Hep G2 Cells , Humans , Methotrexate/chemistry , Methotrexate/pharmacology , Mice, Inbred BALB C , Particle Size , Surface Properties
9.
Mol Pharm ; 12(4): 1318-27, 2015 Apr 06.
Article in English | MEDLINE | ID: mdl-25710590

ABSTRACT

The particle shape of the drug delivery systems had a strong impact on their in vitro and in vivo performance, but there was limited availability of techniques to produce the specific shaped drug carriers. In this article, the novel methotrexate (MTX) decorated MPEG-PLA nanobacillus (MPEG-PLA-MTX NB) was prepared by the self-assembly technique followed by the extrusion through SPG membrane with high N2 pressure for targeted drug delivery, in which Janus-like MTX was not only used as a specific anticancer drug but could also be served as a tumor-targeting ligand. The MPEG-PLA-MTX NBs demonstrated much higher in vitro and in vivo targeting efficiency compared to the MPEG-PLA-MTX nanospheres (MPEG-PLA-MTX NSs) and MPEG-PLA nanospheres (MPEG-PLA NSs). In addition, the MPEG-PLA-MTX NBs also displayed much more excellent in vitro and in vivo antitumor activity than the MPEG-PLA-MTX NSs and free MTX injection. To our knowledge, this work provided the first example of the integration of the shape design (which mediated an early phase tumor accumulation and a late-phase cell internalization) and Janus-faced function (which mediated an early phase active targeting effect and a late-phase anticancer effect) on the basis of nanoscaled drug delivery systems. The highly convergent and cooperative drug delivery strategy opens the door to more drug delivery systems with new shapes and functions for cancer therapy.


Subject(s)
Bacillus , Drug Delivery Systems , Neoplasms/drug therapy , Polymers/chemistry , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Drug Carriers/chemistry , Flow Cytometry , HeLa Cells , Humans , Lactic Acid/chemistry , Methotrexate/administration & dosage , Mice , Nanoparticles/chemistry , Particle Size , Polyesters/chemistry , Polyethylene Glycols/chemistry
10.
Anal Chem ; 86(13): 6262-7, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24873535

ABSTRACT

We report a simple and rapid method for fabricating a surface-enhanced Raman scattering (SERS) substrate, which offers good flexibility, excellent optical transparency, and high SERS activity. Specifically, the SERS substrate (AuNPs/PMMA film) was obtained through self-assembly of gold nanoparticles (AuNPs) on newborn poly(methyl methacrylate) (PMMA) template. The UV-vis spectroscopy analysis and scanning electron microscopy observation revealed that the gold nanoparticles were closely assembled on the flexible and transparent PMMA template. The fabricated AuNPs/PMMA film SERS substrate allowed detection of model molecule, malachite green isothiocyanate, at a concentration as low as 0.1 nM, and exhibited good reproducibility in the SERS measurement. The Raman enhancement factor (EF) of the AuNPs/PMMA film was found to be as high as (2.4 ± 0.3) × 10(7). In addition, measure of residual malachite green on fish surface was carried out, and the result indicated that the AuNPs/PMMA film had great potential in the in situ ultrasensitive detection of analyte on irregular objects.


Subject(s)
Coloring Agents/analysis , Gold/chemistry , Isothiocyanates/analysis , Metal Nanoparticles/chemistry , Polymethyl Methacrylate/chemistry , Rosaniline Dyes/analysis , Spectrum Analysis, Raman/methods , Limit of Detection , Metal Nanoparticles/ultrastructure , Reproducibility of Results , Surface Properties
11.
Anticancer Drugs ; 25(6): 663-72, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24583771

ABSTRACT

Neuropilin-1 (NRP-1) is a nontyrosine kinase coreceptor for semaphorin 3A and the vascular endothelial growth factor involved in tumor angiogenesis, growth, and metastasis and is regarded as a promising target for cancer therapy. In the present study, we investigated the effects of an anti-NRP-1 monoclonal antibody (mAb) that we generated for MCF7 breast cancer cellular adhesion studies. MTT, colony formation, and adhesion assays showed that our anti-NRP-1 mAb dose-dependently inhibited MCF7 proliferation and fibronectin adhesion, leading to a rounded cellular morphology. Further, rhodamine phalloidin stain revealed that fibronectin-dependent formation of actin stress fibers was inhibited by anti-NRP-1 mAb. Immunoprecipitation and western blot showed that anti-NRP-1 mAb treatment inhibited the formation of NRP-1-α5ß1 integrin complexes and suppressed the phosphorylation of focal adhesion kinase and p130cas in MCF7 cells. These findings contribute to further understanding the NRP-1 function in cell adhesion and tumor metastasis. Moreover, our anti-NRP-1 mAb is a prospective drug candidate for tumor treatment.


Subject(s)
Antibodies, Monoclonal/pharmacology , Breast Neoplasms/pathology , Cell Adhesion/drug effects , Crk-Associated Substrate Protein/metabolism , Fibronectins/physiology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Neuropilin-1/metabolism , Actins/metabolism , Antibodies, Monoclonal/isolation & purification , Female , Humans , MCF-7 Cells , Neuropilin-1/immunology , Signal Transduction
12.
Cell Death Dis ; 15(6): 453, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926362

ABSTRACT

Liver regeneration is a complex process involving the crosstalk between parenchymal and non-parenchymal cells, especially macrophages. However, the underlying mechanisms remain incompletely understood. Here, we identify the E3 ubiquitin ligase TRIM26 as a crucial regulator of liver regeneration. Following partial hepatectomy or acute liver injury induced by carbon tetrachloride, Trim26 knockout mice exhibit enhanced hepatocyte proliferation compared to wild-type controls, while adeno-associated virus (AAV)-mediated overexpression of Trim26 reverses the promotional effects. Mechanistically, Trim26 deficiency promotes the recruitment of macrophages to the liver and their polarization towards pro-inflammatory M1 phenotype. These M1 macrophages secrete Wnts, including Wnt2, which subsequently stimulate hepatocyte proliferation through the activation of Wnt/ß-catenin signaling. In hepatocytes, Trim26 knockdown reduces the ubiquitination and degradation of ß-catenin, thereby further enhancing Wnt/ß-catenin signaling. Pharmacological inhibition of Wnt/ß-catenin pathway by ICG-001 or depletion of macrophages by clodronate liposomes diminishes the pro-regenerative effects of Trim26 deficiency. Moreover, bone marrow transplantation experiments provide evidence that Trim26 knockout in myeloid cells alone can also promote liver regeneration, highlighting the critical role of macrophage Trim26 in this process. Taken together, our study uncovers TRIM26 as a negative regulator of liver regeneration by modulating macrophage polarization and Wnt/ß-catenin signaling in hepatocytes, providing a potential therapeutic target for promoting liver regeneration in clinical settings.


Subject(s)
Hepatocytes , Liver Regeneration , Macrophages , Mice, Knockout , Ubiquitin-Protein Ligases , Wnt Signaling Pathway , beta Catenin , Animals , Male , Mice , beta Catenin/metabolism , Cell Polarity , Cell Proliferation , Hepatocytes/metabolism , Liver/metabolism , Liver/pathology , Macrophages/metabolism , Mice, Inbred C57BL , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
13.
ACS Nano ; 18(35): 24219-24235, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39172516

ABSTRACT

Fibrosarcoma, a malignant mesenchymal tumor, is characterized by aggressive invasiveness and a high recurrence rate, leading to poor prognosis. Anthracycline drugs, such as doxorubicin (DOX), represent the frontline chemotherapy for fibrosarcoma, but often exhibit suboptimal efficacy. Recently, exploiting the stimulator of interferon genes (STING)-mediated innate immunity has emerged as a hopeful strategy for cancer treatment. Integrating chemotherapy with immunomodulators in chemo-immunotherapy has shown potential for enhancing treatment outcomes. Herein, we introduce an advanced dendritic cell (DC) nanovaccine, cGAMP@PLGA@CRTM (GP@CRTM), combined with low-dose DOX to enhance fibrosarcoma chemo-immunotherapy. The nanovaccine consists of poly(lactic-co-glycolic acid) (PLGA) nanoparticles encapsulating the STING agonist 2,3-cGAMP (cGAMP@PLGA, GP) as its core, and a calreticulin (CRT) high-expressing fibrosarcoma cell membrane (CRTM) as the shell. Exposing CRT on the vaccine surface aids in recruiting DCs and stimulating uptake, facilitating efficient simultaneous delivery of STING agonists and tumor antigens to DCs. This dual delivery method effectively activates the STING pathway in DCs, triggering sustained immune stimulation. Simultaneously, low-dose DOX reduces chemotherapy-related side effects, directly kills a subset of tumor cells, and increases tumor immunogenicity, thus further amplifying immune therapeutic performance. Hence, these findings demonstrate the potential of DC nanovaccine GP@CRTM as a booster for chemotherapy. Synergistically combining low-dose DOX with the DC nanovaccine emerges as a powerful chemo-immunotherapy strategy, optimizing systemic fibrosarcoma therapy.


Subject(s)
Cancer Vaccines , Dendritic Cells , Doxorubicin , Fibrosarcoma , Nanoparticles , Nucleotides, Cyclic , Dendritic Cells/immunology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Fibrosarcoma/drug therapy , Fibrosarcoma/pathology , Fibrosarcoma/immunology , Fibrosarcoma/therapy , Animals , Doxorubicin/pharmacology , Doxorubicin/chemistry , Mice , Nucleotides, Cyclic/chemistry , Nucleotides, Cyclic/pharmacology , Nanoparticles/chemistry , Cancer Vaccines/immunology , Humans , Membrane Proteins/metabolism , Cell Line, Tumor , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Mice, Inbred C57BL , Immunotherapy , Calreticulin/metabolism , Nanovaccines
14.
Lasers Med Sci ; 28(6): 1541-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23377406

ABSTRACT

Photodynamic therapy (PDT) has been clinically used for liver cancer. The pharmacokinetics of a photosensitizer needs to be monitored so that PDT can be performed at the most favorable time and with the proper dose to increase the cure rate. As mTHPC is a fluorescent compound, we investigate its pharmacokinetics, distribution, and elimination in the rat orthotropic liver cancer model in order to confirm an optimal treatment opportunity of liver cancer PDT. After intravenous administration at a single dose of 300 µg/kg, mTHPC was extracted from tissue homogenates or plasma. Then, mTHPC concentrations were assessed by fluorescence spectroscopy and the data were processed with PK-GRAPH pharmacokinetic procedure. The plasma concentration-time profile of mTHPC showed a short distribution half-life (T½α = 0.082 h) and a relatively longer elimination half-life (T½ß = 28.23 h), which quite fitted with a two-compartment model. The results of mTHPC tissue distributions showed that the highest drug accumulation was in tumor tissue, and successively decreased in liver, heart, spleen, muscle, and skin tissues. The drug distribution ratio of tumor to normal tissue reached the peak at 24 h after mTHPC administration. mTHPC was eliminated at a suitable rate in rat orthotropic liver cancer model, and there was no long-term accumulation of mTHPC in rat tissues. For PDT of orthotropic liver cancer, 24 h after mTHPC intravenous injection may be the optimal treatment time point, which might provide higher clinical efficacy and reduce side effects.


Subject(s)
Liver Neoplasms, Experimental/drug therapy , Mesoporphyrins/therapeutic use , Photochemotherapy , Photosensitizing Agents/therapeutic use , Animals , Carcinoma 256, Walker/drug therapy , Carcinoma 256, Walker/metabolism , Cell Line, Tumor , Half-Life , Injections, Intravenous , Liver Neoplasms, Experimental/metabolism , Male , Mesoporphyrins/administration & dosage , Mesoporphyrins/pharmacokinetics , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/pharmacokinetics , Rats , Rats, Wistar , Spectrometry, Fluorescence , Tissue Distribution
15.
Aging (Albany NY) ; 15(16): 8113-8136, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37595258

ABSTRACT

OBJECTIVE: Pyroptosis is a form of programmed cell death that is essential for immunity. Herein, this study was conducted to uncover the implication of pyroptosis in immunomodulation and tumor microenvironment (TME) in gastric cancer. METHODS: Prognostic pyroptosis-related genes were extracted to identify different pyroptosis phenotypes and pyroptosis genomic phenotypes via unsupervised clustering analysis in the gastric cancer meta-cohort cohort (GSE15459, GSE62254, GSE84437, GSE26253 and TCGA-STAD). The activation of hallmark gene sets was quantified by GSVA and immune cell infiltration was estimated via ssGSEA and CIBERSORT. Through PCA algorithm, pyroptosis score was conducted. The predictors of immune response (TMB and IPS) and genetic mutations were evaluated. The efficacy of pyroptosis score in predicting immune response was verified in two anti-PD-1 therapy cohorts. RESULTS: Three different pyroptosis phenotypes with different prognosis, biological pathways and tumor immune microenvironment were established among 1275 gastric cancer patients, corresponding to three immune phenotypes: immune-inflamed, immune-desert, and immune-excluded. According to the pyroptosis score, patients were separated into high and low pyroptosis score groups. Low pyroptosis score indicated favorable survival outcomes, enhanced immune responses, and increased mutation frequency. Moreover, low pyroptosis score patients displayed more clinical benefits from anti-PD-1 and prolonged survival time. CONCLUSION: Our findings uncovered a nonnegligible role of pyroptosis in immunomodulation and TME multiformity and complicacy in gastric cancer. Quantifying the pyroptosis score in individual tumors may tailor more effective immunotherapeutic strategies.


Subject(s)
Stomach Neoplasms , Humans , Pyroptosis , Immunotherapy , Immunomodulation , Phenotype , Tumor Microenvironment
16.
Adv Healthc Mater ; 12(8): e2202418, 2023 03.
Article in English | MEDLINE | ID: mdl-36459700

ABSTRACT

The hypoxic character of tumors and the poor targeting ability of photosensitizers often limit the efficacy of photodynamic therapy (PDT). In recent years, the discovery of metal nanoenzymes and nanocarriers has improved PDT. Thereby, to improve the effective utilization of photosensitizers and oxygen (O2 ) in tumors, herein, a nanosystem (LS-HB@HvCeO2 -NRP1 mAb, LHCN1) is reported, in which a hollow virus-like cerium oxide (HvCeO2 ) is surface-decorated with tumor-targeting neuropilin-1 monoclonal antibody (NRP1 mAb), and loaded with a photosensitizer (chlorin e6-C-15-ethyl ester, LS-HB). In vitro and in vivo experiments demonstrate that LHCN1 can efficiently accumulate within the tumor sites via the targeting guidance of NRP1 mAb and is then rapidly endocytosed into cells. Furthermore, HvCeO2 with catalase-mimetic activity can decompose the endogenous hydrogen peroxide (H2 O2 ) to promote O2 via the valence transformation between Ce4+ and Ce3+ , relieving tumor hypoxia and improving the PDT efficacy. Upon near-infrared laser irradiation, LS-HB produces large amounts of cytotoxic reactive oxygen species. Moreover, LHCN1 is used in fluorescence/photoacoustic multimodal imaging for in vivo drug localization, and its use in PDT evidently helps inhibit tumor growth with no apparent toxicity to normal tissues. Thus, LHCN1 may provide a promising strategy for precise tumor-specific diagnosis and treatment.


Subject(s)
Nanoparticles , Photochemotherapy , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Photochemotherapy/methods , Oncogenic Viruses , Cell Line, Tumor , Hydrogen Peroxide , Oxygen
17.
Cancer Med ; 12(18): 18460-18469, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37723872

ABSTRACT

BACKGROUND: The surgical approach and prognosis for invasive adenocarcinoma (IAC) and minimally invasive adenocarcinoma (MIA) of the lung differ. However, they both manifest as identical ground-glass nodules (GGNs) in computed tomography images, and no effective method exists to discriminate them. METHODS: We developed and validated a three-dimensional (3D) deep transfer learning model to discriminate IAC from MIA based on CT images of GGNs. This model uses a 3D medical image pre-training model (MedicalNet) and a fusion model to build a classification network. Transfer learning was utilized for end-to-end predictive modeling of the cohort data of the first center, and the cohort data of the other two centers were used as independent external validation data. This study included 999 lung GGN images of 921 patients pathologically diagnosed with IAC or MIA at three cohort centers. RESULTS: The predictive performance of the model was assessed using the area under the receiver operating characteristic curve (AUC). The model had high diagnostic efficacy for the training and validation groups (accuracy: 89%, sensitivity: 95%, specificity: 84%, and AUC: 95% in the training group; accuracy: 88%, sensitivity: 84%, specificity: 93%, and AUC: 92% in the internal validation group; accuracy: 83%, sensitivity: 83%, specificity: 83%, and AUC: 89% in one external validation group; accuracy: 78%, sensitivity: 80%, specificity: 77%, and AUC: 82% in the other external validation group). CONCLUSIONS: Our 3D deep transfer learning model provides a noninvasive, low-cost, rapid, and reproducible method for preoperative prediction of IAC and MIA in lung cancer patients with GGNs. It can help clinicians to choose the optimal surgical strategy and improve the prognosis of patients.

18.
Front Oncol ; 13: 1078863, 2023.
Article in English | MEDLINE | ID: mdl-36890815

ABSTRACT

Background: This study aimed to establish an effective model for preoperative prediction of tumor deposits (TDs) in patients with rectal cancer (RC). Methods: In 500 patients, radiomic features were extracted from magnetic resonance imaging (MRI) using modalities such as high-resolution T2-weighted (HRT2) imaging and diffusion-weighted imaging (DWI). Machine learning (ML)-based and deep learning (DL)-based radiomic models were developed and integrated with clinical characteristics for TD prediction. The performance of the models was assessed using the area under the curve (AUC) over five-fold cross-validation. Results: A total of 564 radiomic features that quantified the intensity, shape, orientation, and texture of the tumor were extracted for each patient. The HRT2-ML, DWI-ML, Merged-ML, HRT2-DL, DWI-DL, and Merged-DL models demonstrated AUCs of 0.62 ± 0.02, 0.64 ± 0.08, 0.69 ± 0.04, 0.57 ± 0.06, 0.68 ± 0.03, and 0.59 ± 0.04, respectively. The clinical-ML, clinical-HRT2-ML, clinical-DWI-ML, clinical-Merged-ML, clinical-DL, clinical-HRT2-DL, clinical-DWI-DL, and clinical-Merged-DL models demonstrated AUCs of 0.81 ± 0.06, 0.79 ± 0.02, 0.81 ± 0.02, 0.83 ± 0.01, 0.81 ± 0.04, 0.83 ± 0.04, 0.90 ± 0.04, and 0.83 ± 0.05, respectively. The clinical-DWI-DL model achieved the best predictive performance (accuracy 0.84 ± 0.05, sensitivity 0.94 ± 0. 13, specificity 0.79 ± 0.04). Conclusions: A comprehensive model combining MRI radiomic features and clinical characteristics achieved promising performance in TD prediction for RC patients. This approach has the potential to assist clinicians in preoperative stage evaluation and personalized treatment of RC patients.

19.
Oncol Res ; 32(2): 361-371, 2023.
Article in English | MEDLINE | ID: mdl-38186571

ABSTRACT

The high mortality rate associated with gastric cancer (GC) has resulted in an urgent need to identify novel therapeutic targets for GC. This study aimed to investigate whether GAIP interacting protein, C terminus 1 (GIPC1) represents a therapeutic target and its regulating mechanism in GC. GIPC1 expression was elevated in GC tissues, liver metastasis tissues, and lymph node metastases. GIPC1 knockdown or GIPC1 blocking peptide blocked the platelet-derived growth factor receptor (PDGFR)/PI3K/AKT signaling pathway, and inhibited the proliferation and migration of GC cells. Conversely, GIPC1 overexpression markedly activated the PDGFR/PI3K/AKT signaling pathway, and promoted GC cell proliferation and migration. Furthermore, platelet-derived growth factor subunit BB (PDGF-BB) cytokines and the AKT inhibitor attenuated the effect of differential GIPC1 expression. Moreover, GIPC1 silencing decreased tumor growth and migration in BALB/c nude mice, while GIPC1 overexpression had contrasting effects. Taken together, our findings suggest that GIPC1 functions as an oncogene in GC and plays a central role in regulating cell proliferation and migration via the PDGFR/PI3K/AKT signaling pathway.


Subject(s)
Stomach Neoplasms , Humans , Animals , Mice , Stomach Neoplasms/genetics , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Mice, Nude , Signal Transduction , Adaptor Proteins, Signal Transducing
20.
Adv Sci (Weinh) ; 10(30): e2303872, 2023 10.
Article in English | MEDLINE | ID: mdl-37661565

ABSTRACT

The lethality and chemotherapy resistance of pancreatic cancer necessitates the urgent development of innovative strategies to improve patient outcomes. To address this issue, we designed a novel drug delivery system named GDMCN2,which uses iron-based metal organic framework (Fe-MOF) nanocages encased in a covalent organic framework (COF) and modified with the pancreatic cancer-specific antibody, NRP2. After being targeted into tumor cells, GDMCN2 gradually release the sonosensitizer sinoporphyrin sodium (DVDMS) and chemotherapeutic gemcitabine (GEM) and simultaneously generated reactive oxygen species (ROS) under ultrasound (US) irradiation. This system can overcome gemcitabine resistance in pancreatic cancer and reduce its toxicity to non-targeted cells and tissues. In a mechanistic cascade, the release of ROS activates the mitochondrial transition pore (MPTP), leading to the release of Ca2+ and induction of endoplasmic reticulum (ER) stress. Therefore, microtubule-associated protein 1A/1B-light chain 3 (LC3) is activated, promoting lysosomal autophagy. This process also induces autophagy-dependent ferroptosis, aided by the upregulation of Nuclear Receptor Coactivator 4 (NCOA4). This mechanism increases the sensitivity of pancreatic cancer cells to chemotherapeutic drugs and increases mitochondrial and DNA damage. The findings demonstrate the potential of GDMCN2 nanocages as a new avenue for the development of cancer therapeutics.


Subject(s)
Ferroptosis , Metal-Organic Frameworks , Pancreatic Neoplasms , Humans , Metal-Organic Frameworks/metabolism , Cell Line, Tumor , Reactive Oxygen Species/metabolism , Apoptosis , Antibodies, Monoclonal/therapeutic use , Autophagy , Gemcitabine , Pancreatic Neoplasms/drug therapy , Endoplasmic Reticulum/metabolism , Pancreatic Neoplasms
SELECTION OF CITATIONS
SEARCH DETAIL