Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
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
2.
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.

3.
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
4.
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.

5.
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
6.
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
8.
J Mol Histol ; 53(5): 781-791, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35920984

ABSTRACT

Aldo-keto reductase family one, member B10 (AKR1B10) has been reported to be involved in the tumorigenesis of various cancers. It has been reported that colorectal cancer is closely associated with chronic inflammation, but the underlying molecular mechanisms are still elusive. In our study, we evaluated the relationship between AKR1B10 expression and clinicopathological characteristics of colon cancer and showed that AKR1B10 expression was significantly correlated with the T stage and clinical stage of colon cancer. Knockdown of AKR1B10 significantly decreased the expression of the inflammatory cytokines IL1α and IL6 induced by lipopolysaccharide by inhibiting the NF-κB signaling pathway. Furthermore, AKR1B10 depends on its reductase activity to affect the NF-κB signaling pathway and subsequently affect the production of inflammatory cytokines. In addition, knockdown of AKR1B10 effectively reduced cell proliferation and clonogenic growth, indicating the biological role of AKR1B10 in colon cancer. Together, our findings provide important insights into a previously unrecognized role of AKR1B10 in colon cancer.


Subject(s)
Aldo-Keto Reductases , Colonic Neoplasms , Cytokines , NF-kappa B , Signal Transduction , Aldo-Keto Reductases/metabolism , Cell Line, Tumor , Colonic Neoplasms/genetics , Cytokines/metabolism , Humans , NF-kappa B/metabolism
9.
Adv Healthc Mater ; 11(16): e2102770, 2022 08.
Article in English | MEDLINE | ID: mdl-35575205

ABSTRACT

The self-delivery of sonosensitizers and immunomodulators to tumor areas, which is highly recommended for enhancing sonodynamic immunotherapy, remains a challenge. Herein, a self-delivering nanodrug (HB-NLG8189, drug loading: ≈100 wt%) is developed by the small-molecule self-assembly of "HB" (a new clinical photosensitizer) and NLG8189 (indoleamine-(2,3)-dioxygenase (IDO) pathway inhibitor) for sonodynamic-augmented immunotherapy; this preparation method ensures the absence of excipient-related toxicity and immunogenicity. To evade immune recognition and prolong the circulation time, the HB-NLG8189 nanodrugs are camouflaged using macrophage cell membranes (MPCMs). The constructed HB-NLG8189@MPCM nanodrugs show an ability to preferentially accumulate within tumors. Upon ultrasound triggering, the HB-NLG8189@MPCM is able to generate reactive oxygen species efficiently for robust sonodynamic therapy; it induces immunogenic cell death, initiates an antitumor immune response to activate tumor-specific effector T cells, and promotes the secretion of inflammatory cytokines. The concomitant delivery of NLG8189 reverses the immunosuppressive tumor microenvironment by restraining IDO-1 activation and the intratumoral infiltration of regulatory T cells. Sonodynamic-augmented immunotherapy with HB-NLG8189@MPCM significantly inhibits the growth of both primary and distant tumors with little systemic toxicity. The biomimetic self-delivery nanodrug provides a promising paradigm for improving sonodynamic immunotherapy.


Subject(s)
Nanoparticles , Neoplasms , Cell Line, Tumor , Humans , Immunologic Factors/therapeutic use , Immunotherapy , Indoleamine-Pyrrole 2,3,-Dioxygenase , Macrophages/pathology , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology , Tumor Microenvironment
10.
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
11.
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
13.
Adv Healthc Mater ; 11(6): e2102060, 2022 03.
Article in English | MEDLINE | ID: mdl-34894092

ABSTRACT

The anti-tumor efficacy of single photodynamic therapy (PDT) and radiotherapy (RT) has been greatly affected by inadequate tumor uptake of photo/radiation sensitizers, limited laser penetration depth, and radiation sickness caused by high doses of X-rays. Here, the authors report a biomimetic coronavirus-inspired hollow mesoporous gadolinium/bismuth nanocarrier loaded with a new NIR photosensitizer HB (termed as HB@VHMBi-Gd) for magnetic resonance imaging (MRI)-guided synergistic photodynamic-RT. HB@VHMBi-Gd displayed a faster cellular uptake rate than the conventional spherical HMBi-Gd loaded with HB (HB@SHMBi-Gd) because of rough surface-enhanced adhesion. After intravenous injection, HB@VHMBi-Gd is efficiently delivered to the tumor and rapidly invades the tumor cells by surface spikes. Interestingly, lysosomal acidity can trigger the degradation of VHMBi-Gd to produce ultrasmall nanoparticles to amplify the X-ray attenuation ability and enhance MRI contrast and radiosensitization. Under laser and X-ray irradiation, HB@VHMBi-Gd significantly enhances 1 O2 generation from HB to induce activation of caspase 9/3 and inhibition of C-myc, while enhancing hydroxyl radical generation from Bi2 O3 to induce intense DNA breakage. By synergistically inducing cell apoptosis by distinct reactive oxygen species (ROS), HB@VHMBi-Gd exhibits superior anticancer efficacy with ≈90% tumor inhibition. They envision that biomimetic virus-inspired hollow hybrid metal nanoparticles can provide a promising strategy for imaging-guided synergistic photodynamic-RT.


Subject(s)
Metal Nanoparticles , Nanoparticles , Photochemotherapy , Bismuth , Cell Line, Tumor , Gadolinium , Magnetic Resonance Imaging , Photochemotherapy/methods , Theranostic Nanomedicine/methods
14.
ACS Appl Mater Interfaces ; 13(37): 44013-44027, 2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34494427

ABSTRACT

Synergetic therapy includes the combination of two or more conventional therapeutic approaches and can be used for tumor treatment by combining the advantages and avoiding the drawbacks of each type of treatment. In the present study, truncated tissue factor (tTF)-EG3287 fusion protein-encapsulated gold nanorod (GNR)-virus-inspired mesoporous silica core-shell nanoparticles (vinyl hybrid silica nanoparticles; VSNP) (GNR@VSNP-tTF-EG3287) were synthesized to achieve synergetic therapy by utilizing selective vascular thrombosis therapy (SVTT) and photothermal therapy (PTT). By integrating the targeted coagulation activity of tTF-EG3287 and the high tumor ablation effect of GNR@VSNP, local hyperthermia could induce a high percentage of apoptosis of vascular endothelial cells by using near-infrared light. This provided additional phospholipid sites for tTF-EG3287 and enhanced its procoagulant activity in vitro. In addition, the nanoparticles, which had unique topological viral structures, exhibited superior cellular uptake properties leading to significant antitumor efficacy. The in vivo antitumor results further demonstrated an interaction between SVTT and PTT, whereas the synergetic therapy (SVTT and PTT) achieved an enhanced effect, which was superior to the respective treatment efficacy of each modality or the additive effect of their individual efficacies. In summary, the synthesized GNR@VSNP-tTF-EG3287 exerted synergetic effects and enhanced the antitumor efficiency by avoiding multiple injections and suboptimal administration. These effects simultaneously affected both tumor blood supply and cancer cell proliferation. The data suggested that the integration of SVTT induced by tTF-EG3287 and PTT could provide potential strategies for synergetic tumor therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Coagulants/therapeutic use , Nanotubes/chemistry , Neoplasms/drug therapy , Recombinant Fusion Proteins/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Coagulants/chemistry , Female , Gold/chemistry , Gold/radiation effects , Gold/toxicity , Hep G2 Cells , Human Umbilical Vein Endothelial Cells , Humans , Infrared Rays , Mice, Inbred BALB C , Mice, Nude , Nanotubes/radiation effects , Nanotubes/toxicity , Peptide Fragments/chemistry , Peptide Fragments/therapeutic use , Photothermal Therapy , Porosity , Rabbits , Recombinant Fusion Proteins/chemistry , Silicon Dioxide/chemistry , Silicon Dioxide/radiation effects , Silicon Dioxide/toxicity , Thromboplastin/chemistry , Thromboplastin/therapeutic use , Thrombosis/chemically induced , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/therapeutic use , Xenograft Model Antitumor Assays
15.
Drug Deliv ; 28(1): 1769-1784, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34470548

ABSTRACT

Traditional combined photodynamic and photothermal therapy (PDT/PTT) was limited in clinical treatment of cancer due to the exceptionally low drug delivery efficiency to tumor sites and the activation by laser excitation with different wavelengths. We have accidentally discovered that our synthesized chlorin e6-C-15-ethyl ester (HB, a new type of photosensitizer) be activated by a laser with an excitation wavelength of 660 nm. Herein, we utilized Au nanorods (AuNRs) as 660 nm-activated PTT carriers to be successively surface-functionalized with HB and tumor-targeting peptide cyclic RGD (cRGD) to develop HB-AuNRs@cRGD for single NIR laser-induced targeted PDT/PTT. The HB-AuNRs@cRGD could be preferentially accumulated within tumor sites and rapidly internalized by cancer cells. Thereby, the HB-AuNRs@cRGD could exhibit amplified therapeutic effects by producing both significant reactive oxygen species (ROS) and hyperthermia simultaneously under the guidance of fluorescence imaging. The tumor inhibition rate on ECA109 esophageal cancer model was approximately 77.04%, and the negligible systematic toxicity was observed. This study proposed that HB-AuNRs@cRGD might be a promising strategy for single NIR laser-induced and imaging-guided targeted bimodal phototherapy.


Subject(s)
Gold/chemistry , Nanotubes/chemistry , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Photothermal Therapy/methods , Animals , Cell Line , Female , Low-Level Light Therapy , Mice , Mice, Inbred BALB C , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/pharmacokinetics , Reactive Oxygen Species/metabolism
16.
Front Oncol ; 11: 657008, 2021.
Article in English | MEDLINE | ID: mdl-34336654

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy with extremely limited treatment; the effective targeting strategy stays an urgent unmet need. Neuropilin-2 (NRP2), a multifunctional transmembrane non-tyrosine-kinase glycoprotein, enhances various signal transduction pathways to modulate cancer progression. However, the application value of NRP2 as a therapeutic target in pancreatic cancer is still unclear. Here, we detected the elevated NRP2 was associated with the poor prognosis of pancreas carcinoma. The mouse monoclonal antibody targeting NRP2 (N2E4) that could specifically bind to PDAC cells was developed. Moreover, N2E4 inhibits PDAC proliferation, migration, and invasion in vitro, and repressed growth and metastasis in vivo. Mechanistically, the effect of N2E4 was mainly related to the blocking of interaction between NRP2 with integrinß1 to inhibit FAK/Erk/HIF-1a/VEGF signaling. Therefore, N2E4 has the potential for targeting therapy of PDAC. This study lays a foundation for the future development of NRP2-based targeted therapy for PDAC.

17.
Nanoscale Res Lett ; 16(1): 131, 2021 Aug 14.
Article in English | MEDLINE | ID: mdl-34390420

ABSTRACT

X-ray computed tomography (CT) has been widely used in clinical practice, and contrast agents such as Iohexol are often used to enhance the contrast of CT imaging between normal and diseased tissue. However, such contrast agents can have some toxicity. Thus, new CT contrast agents are urgently needed. Owing to the high atomic number (Z = 83), low cost, good biological safety, and great X-ray attenuation property (5.74 cm2 kg-1 at 100 keV), bismuth has gained great interest from researchers in the field of nano-sized CT contrast agents. Here, we synthesized BiF3: Ln@PVP nanoparticles (NPs) with an average particle size of about 380 nm. After coating them with polyvinylpyrrolidone (PVP), the BiF3: Ln@PVP NPs possessed good stability and great biocompatibility. Meanwhile, compared with the clinical contrast agent Iohexol, BiF3: Ln@PVP NPs showed superior in vitro CT imaging contrast. Subsequently, after in situ injection with BiF3: Ln@PVP NPs, the CT value of the tumor site after the injection was significantly higher than that before the injection (the CT value of the pre-injection and post-injection was 48.9 HU and 194.58 HU, respectively). The morphology of the gastrointestinal (GI) tract can be clearly observed over time after oral administration of BiF3: Ln@PVP NPs. Finally, the BiF3: Ln@PVP NPs were completely discharged from the GI tract of mice within 48 h of oral administration with no obvious damage to the GI tract. In summary, our easily synthesized BiF3: Ln@PVP NPs can be used as a potential clinical contrast agent and may have broad application prospects in CT imaging.

18.
ACS Omega ; 6(16): 10723-10734, 2021 Apr 27.
Article in English | MEDLINE | ID: mdl-34056226

ABSTRACT

High-efficiency nanotheranostic agents with multimodal imaging guidance have attracted considerable interest in the field of cancer therapy. Herein, novel silver-decorated bismuth-based heterostructured polyvinyl pyrrolidone nanoparticles (NPs) with good biocompatibility (Bi-Ag@PVP NPs) were synthesized for accurate theranostic treatment, which can integrate computed tomography (CT)/photoacoustic (PA) imaging and photodynamic therapy/photothermal therapy (PDT/PTT) into one platform. The Bi-Ag@PVP NPs can enhance light absorption and achieve a better photothermal effect than bismuth NPs. Moreover, after irradiation under an 808 nm laser, the Bi-Ag@PVP NPs can efficiently induce the generation of reactive oxygen species (ROS), thereby synergizing PDT/PTT to exert an efficient tumor ablation effect both in vitro and in vivo. Furthermore, Bi-Ag@PVP NPs can also be employed to perform enhanced CT/PA imaging because of their high X-ray absorption attenuation and enhanced photothermal conversion. Thus, they can be utilized as a highly effective CT/PA imaging-guided nanotheranostic agent. In addition, an excellent antibacterial effect was achieved. After irradiation under an 808 nm laser, the Bi-Ag@PVP NPs can destroy the integrity of Escherichia coli, thereby inhibiting E. coli growth, which can minimize the risk of infection during cancer therapy. In conclusion, our study provides a novel nanotheranostic platform that can achieve CT/PA-guided PDT/PTT synergistic therapy and have potential antibacterial properties. Thus, this work provides an effective strategy for further broad clinical application prospects.

19.
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
20.
Cell Mol Gastroenterol Hepatol ; 10(3): 545-559, 2020.
Article in English | MEDLINE | ID: mdl-32447051

ABSTRACT

BACKGROUND & AIMS: Transforming growth factor (TGF-ß)-induced activation of quiescent hepatic stellate cells (HSCs) and their transformation to myofibroblasts is a key event in liver fibrosis and portal hypertension. GIPC (also referred to as synectin) is a downstream signal activation molecule of TGF-ß and other receptors. In this study, we sought to identify novel genes targeted by TGF-ß and GIPC and elucidate if and how they may contribute to liver fibrosis. METHODS: We performed sequential messenger RNA sequencing analysis on TGF-ß-stimulated HSCs and then on TGF-ß-stimulated HSCs in the presence and absence of GIPC also referred to as synectin (GIPC) knockdown. Insulin-like growth factor binding protein-3 (IGFBP-3) transport protein emerged as a top activation target of both TGF-ß and GIPC. Quantitative polymerase chain reaction, enzyme-linked immunosorbent assay, targeted chromatin immunoprecipitation, and Western blot analysis were done for further confirmation. RESULTS: IGFBP-3, an insulin growth factor transport protein, emerged as a top activation target of both TGF-ß and GIPC, which was confirmed by quantitative polymerase chain reaction, enzyme-linked immunosorbent assay, and Western blot analysis. Targeted chromatin immunoprecipitation showed that GIPC increases the histone 3 lysine 27 (H3K27) acetylation activating mark and concurrently decreases the H3K27 inhibitory trimethylation (H3K27m3) mark, providing an epigenetic correlate to the gene regulation changes. In vivo, global knockout of IGFBP-3 mice resulted in attenuation of HSC activation markers and attenuation of portal pressure in response to chronic liver injury models. Analysis of serum levels from cirrhotic patients also showed an IGFBP-3 increase of more than 2-fold compared with healthy controls. Finally, in vitro mechanism studies showed that IGFBP-3 promotes HSC migration through integrin-dependent phosphorylation of protein kinase B. CONCLUSIONS: TGF-ß up-regulates IGFBP-3 through GIPC, leading to increased HSC migration in vitro and promotes portal hypertension in vivo. These studies support the role of IGFBP-3 as a potential pathophysiologic target or biomarker in chronic liver disease.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Hypertension, Portal/immunology , Insulin-Like Growth Factor Binding Protein 3/metabolism , Integrin beta1/metabolism , Liver Cirrhosis/immunology , Transforming Growth Factor beta/metabolism , Acetylation , Adaptor Proteins, Signal Transducing/genetics , Animals , Biomarkers/blood , Biomarkers/metabolism , Cell Movement/immunology , Cells, Cultured , Disease Models, Animal , Epigenesis, Genetic/immunology , Gene Knockdown Techniques , Hepatic Stellate Cells/immunology , Hepatic Stellate Cells/pathology , Histones/metabolism , Humans , Hypertension, Portal/pathology , Insulin-Like Growth Factor Binding Protein 3/blood , Liver Cirrhosis/pathology , Mice , Mice, Knockout , Phosphorylation/immunology , Primary Cell Culture , Signal Transduction/immunology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...