Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Dig Dis Sci ; 69(5): 1852-1862, 2024 May.
Article in English | MEDLINE | ID: mdl-38514499

ABSTRACT

BACKGROUND: Early rebleeding is a significant complication of endoscopic treatment for esophagogastric variceal hemorrhage (EGVH). However, a reliable predictive model is currently lacking. AIMS: To identify risk factors for rebleeding within 6 weeks and establish a nomogram for predicting early rebleeding after endoscopic treatment of EVGH. METHODS: Demographic information, comorbidities, preoperative evaluation, endoscopic features, and laboratory tests were collected from 119 patients who were first endoscopic treatment for EGVH. Independent risk factors for early rebleeding were determined through least absolute shrinkage and selection operator logistic regression. The discrimination, calibration, and clinical utility of the nomogram were assessed and compared with the model for end-stage liver disease (MELD), Child-Pugh, and albumin-bilirubin (ALBI) scores using receiver-operating characteristic (ROC) curves, calibration plots, and decision curve analyses (DCA). RESULTS: Early rebleeding occurred in 39 patients (32.8%) within 6 weeks after endoscopic treatment. Independent early rebleeding factors included gastric variceal hemorrhage (GVH), concomitant hepatocellular carcinoma (HCC), international normalized ratio (INR), and creatinine. The nomogram demonstrated exceptional calibration and discrimination capability. The area under the curve for the nomogram was 0.758 (95% CI 0.668-0.848), and it was validated at 0.71 through cross-validation and bootstrapping validation. The DCA and ROC curves demonstrated that the nomogram outperformed the MELD, Child-Pugh, and ALBI scores. CONCLUSIONS: Compared with existing prediction scores, the nomogram demonstrated superior discrimination, calibration, and clinical applicability for predicting rebleeding in patients with EGVH after endoscopic treatment. Therefore, it may assist clinicians in the early implementation of aggressive treatment and follow-up.


Subject(s)
Esophageal and Gastric Varices , Gastrointestinal Hemorrhage , Nomograms , Recurrence , Humans , Esophageal and Gastric Varices/surgery , Esophageal and Gastric Varices/diagnosis , Esophageal and Gastric Varices/etiology , Male , Female , Middle Aged , Gastrointestinal Hemorrhage/etiology , Gastrointestinal Hemorrhage/surgery , Gastrointestinal Hemorrhage/diagnosis , Aged , Risk Factors , Retrospective Studies , ROC Curve , Predictive Value of Tests , Adult
2.
Mol Cancer ; 22(1): 21, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36721170

ABSTRACT

BACKGROUND: Excessive extracellular matrix deposition and increased stiffness are typical features of solid tumors such as hepatocellular carcinoma (HCC) and pancreatic ductal adenocarcinoma (PDAC). These conditions create confined spaces for tumor cell migration and metastasis. The regulatory mechanism of confined migration remains unclear. METHODS: LC-MS was applied to determine the differentially expressed proteins between HCC tissues and corresponding adjacent tissue. Collective migration and single cell migration microfluidic devices with 6 µm-high confined channels were designed and fabricated to mimic the in vivo confined space. 3D invasion assay was created by Matrigel and Collagen I mixture treat to adherent cells. 3D spheroid formation under various stiffness environment was developed by different substitution percentage GelMA. Immunoprecipitation was performed to pull down the LH1-binding proteins, which were identified by LC-MS. Immunofluorescent staining, FRET, RT-PCR, Western blotting, FRAP, CCK-8, transwell cell migration, wound healing, orthotopic liver injection mouse model and in vivo imaging were used to evaluate the target expression and cellular phenotype. RESULTS: Lysyl hydroxylase 1 (LH1) promoted the confined migration of cancer cells at both collective and single cell levels. In addition, LH1 enhanced cell invasion in a 3D biomimetic model and spheroid formation in stiffer environments. High LH1 expression correlated with poor prognosis of both HCC and PDAC patients, while it also promoted in vivo metastasis. Mechanistically, LH1 bound and stabilized Septin2 (SEPT2) to enhance actin polymerization, depending on the hydroxylase domain. Finally, the subpopulation with high expression of both LH1 and SEPT2 had the poorest prognosis. CONCLUSIONS: LH1 promotes the confined migration and metastasis of cancer cells by stabilizing SEPT2 and thus facilitating actin polymerization.


Subject(s)
Carcinoma, Hepatocellular , Carcinoma, Pancreatic Ductal , Liver Neoplasms , Pancreatic Neoplasms , Animals , Mice , Actins , Carcinoma, Hepatocellular/genetics , Carcinoma, Pancreatic Ductal/genetics , Liver Neoplasms/genetics , Pancreatic Neoplasms/genetics , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/genetics , Septins
3.
Cancer Sci ; 114(3): 793-805, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36369883

ABSTRACT

Sorafenib is one a first-line therapeutic drugs for advanced hepatocellular carcinoma (HCC). However, only 30% of patients benefit from sorafenib due to drug resistance. We and other groups have revealed that nuclear factor I B (NFIB) regulates liver regeneration and carcinogenesis, but its role in drug resistance is poorly known. We found that NFIB was more upregulated in sorafenib-resistant SMMC-7721 cells compared to parental cells. NFIB knockdown not only sensitized drug-resistant cells to sorafenib but also inhibited the proliferation and invasion of these cells. Meanwhile, NFIB promoted the proliferation and invasion of HCC cells in vitro and facilitated tumor growth and metastasis in vivo. Knocking down NFIB synergetically inhibited tumor growth with sorafenib. Mechanically, gene expression profiling and subsequent verification experiments proved that NFIB could bind with the promoter region of a complex I inhibitor NDUFA4L2 and promote its transcription. Transcriptional upregulation of NDUFA4L2 by NFIB could thus inhibit the sorafenib-induced reactive oxygen species accumulation. Finally, we found that NFIB was highly expressed in HCC tissues, and high NFIB expression level was associated with macrovascular invasion, advanced tumor stage, and poor prognosis of HCC patients (n = 156). In summary, we demonstrated that NFIB could transcriptionally upregulate NDUFA4L2 to enhance both intrinsic and acquired sorafenib resistance of HCC cells by reducing reactive oxygen species induction.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/pathology , NFI Transcription Factors/genetics , Reactive Oxygen Species/metabolism , Sorafenib/pharmacology
4.
Small ; 19(16): e2207194, 2023 04.
Article in English | MEDLINE | ID: mdl-36634971

ABSTRACT

Cancer metastasis is the major cause of cancer-related death. Excessive extracellular matrix deposition and increased stiffness are typical features of solid tumors, creating confined spaces for tumor cell migration and metastasis. Confined migration is involved in all metastasis steps. However, confined and unconfined migration inhibitors are different and drugs available to inhibit confined migration are rare. The main challenges are the modeling of confined migration, the suffering of low throughput, and others. Microfluidic device has the advantage to reduce reagent consumption and enhance throughput. Here, a microfluidic chip that can achieve multi-function drug screening against the collective migration of cancer cells under confined environment is designed. This device is applied to screen out effective drugs on confined migration among a novel mechanoreceptors compound library (166 compounds) in hepatocellular carcinoma, non-small lung cancer, breast cancer, and pancreatic ductal adenocarcinoma cells. Three compounds that can significantly inhibit confined migration in pan-cancer: mitochonic acid 5 (MA-5), SB-705498, and diphenyleneiodonium chloride are found. Finally, it is elucidated that these drugs targeted mitochondria, actin polymerization, and cell viability, respectively. In sum, a high-throughput microfluidic platform for screening drugs targeting confined migration is established and three novel inhibitors of confined migration in multiple cancer types are identified.


Subject(s)
Lung Neoplasms , Microfluidic Analytical Techniques , Humans , Drug Evaluation, Preclinical , Cell Movement , Microfluidics , Lab-On-A-Chip Devices
5.
J Transl Med ; 21(1): 684, 2023 10 02.
Article in English | MEDLINE | ID: mdl-37784113

ABSTRACT

BACKGROUND: There is little prospective evidence exists about whether adherence to a diabetes risk reduction diet (DRRD) is related to a significant reduction in renal cancer risk. We sought to clarify whether adherence to DRRD was associated with a reduced risk of renal cancer in a US population. METHODS: A population-based cohort of 101,755 American adults was identified from the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. A DRRD score was calculated to assess adherence to this dietary pattern, where increased scores indicated greater adherence. The relationship between DRRD score and risk of renal cancer was assessed based on the hazard ratios (HRs) and 95% confidence intervals (CIs), which were both calculated using Cox regression. Non-linear association was determined through restricted cubic spline regression. Potential effect modifiers were identified through subgroup analyses. RESULTS: Over a mean follow-up of 8.8 years, 446 renal cancers were detected. In this analysis, the fully adjusted model depicted a notable 29% reduction in the risk of renal cancer among individuals in the highest quartile of DRRD score in comparison with the lowest quartile individuals (HRQ4 vs. Q1: 0.71; 95% CI = 0.54, 0.94; Ptrend = 0.008). This association remained consistent across a series of sensitivity analyses. A non-linear inverse dose-response association between renal cancer risk with DRRD score was observed (Pnonlinearity = 0.026). Subgroup analyses showed that this favorable link was more prominent in participants with low Healthy Eating Index-2015 (Pinteraction = 0.015). Regarding the individual components of DRRD, a decrease in the risk of renal cancer was linked to increased intake of cereal fiber and whole fruit, and lower sugar-sweetened beverage consumption (all Ptrend < 0.05). CONCLUSIONS: Our findings indicate that individuals adhering to DRRD are associated with a reduction in the risk of renal cancer.


Subject(s)
Diabetes Mellitus , Kidney Neoplasms , Male , Adult , Humans , United States/epidemiology , Prospective Studies , Diet , Diet, Reducing , Kidney Neoplasms/epidemiology , Risk Reduction Behavior , Risk Factors
6.
Cancer Sci ; 113(2): 478-488, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34826159

ABSTRACT

The aim of this study was to: (a) explore the potential mechanism of cancer cell sensitivity to cisplatin, docetaxel, and 5-fluorouracil (TPF) in oral squamous cell carcinoma (OSCC) patients overexpressing growth differentiation factor 15 (GDF15); and (b) identify potential alternative agents for patients who might not benefit from inductive TPF chemotherapy. The results indicated that OSCC cells overexpressing GDF15 were sensitive to TPF through a caspase-9-dependent pathway both in vitro and in vivo. Immunoprecipitation combined with mass spectrometry revealed that the erbB2 protein was a potential GDF15-binding protein, which was verified by coimmunoprecipitation. Growth differentiation factor 15 overexpression promoted OSCC cell proliferation through erbB2 phosphorylation, as well as downstream AKT and Erk signaling pathways. When GDF15 expression was blocked, the phosphorylation of both the erbB2 and AKT/Erk pathways was downregulated. When OSCC cells with GDF15 overexpression were treated with the erbB2 phosphorylation inhibitor, CI-1033, cell proliferation and xenograft growth colony formation were significantly blocked (P < .05). Thus, GDF15-overexpressing OSCC tumors are sensitive to TPF chemoagents through caspase-9-dependent pathways. Growth differentiation factor 15 overexpression promotes OSCC proliferation through erbB2 phosphorylation. Thus, ErbB2 inhibitors could represent potential targeted drugs or an alternative therapy for OSCC patients with GDF15 overexpression.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Growth Differentiation Factor 15/metabolism , Mouth Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Squamous Cell Carcinoma of Head and Neck/metabolism , Animals , Apoptosis , Caspase 9/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cisplatin/pharmacology , Fluorouracil/pharmacology , Humans , Mice , Morpholines/pharmacology , Phosphorylation/drug effects , Receptor, ErbB-2/antagonists & inhibitors , Signal Transduction/drug effects , Taxoids/pharmacology
7.
BMC Oral Health ; 22(1): 265, 2022 06 29.
Article in English | MEDLINE | ID: mdl-35768820

ABSTRACT

BACKGROUND: Schwannomas or neurilemmomas are well-encapsulated, benign, solitary, and slow-growing tumors that originate from Schwann cells of the nerve sheath. Extracranial schwannoma is reported to have a relatively high incidence in the tongue while an extremely low incidence in the floor of mouth. In the current study, we presented the first case series of hypoglossal nerve-derived schwannoma in the floor of mouth in Asia. METHODS: A retrospective study of 9 surgical cases of hypoglossal nerve-derived schwannoma in the floor of mouth was performed. The patient and tumor characteristics were evaluated by physical, radiological and pathological examination. Details of operation and complications were also recorded. RESULTS: Hypoglossal nerve-derived schwannoma in the floor of mouth showed a well-defined boundary with a firm texture, smooth surface and good mobility on palpation. The median maximum diameter of the tumors was 4.3 cm (range 2.8-7.0 cm). The median operative time and bleeding volumes were 89.4 min (range 47-180 min) and 99.2 mL (range 15-200 mL), respectively. All cases received complete surgical excision. CONCLUSION: In this study, we presented the diagnosis and management of hypoglossal nerve-derived schwannoma in the floor of mouth for the first time in Asia. The study provided us with a recommendation for consideration of the diagnosis of hypoglossal schwannoma when a patient presents with a mass in the floor of mouth.


Subject(s)
Cranial Nerve Neoplasms , Hypoglossal Nerve Diseases , Neurilemmoma , Cranial Nerve Neoplasms/diagnosis , Cranial Nerve Neoplasms/pathology , Cranial Nerve Neoplasms/surgery , Humans , Hypoglossal Nerve/pathology , Hypoglossal Nerve/surgery , Hypoglossal Nerve Diseases/diagnosis , Hypoglossal Nerve Diseases/etiology , Hypoglossal Nerve Diseases/surgery , Mouth Floor/pathology , Mouth Floor/surgery , Neurilemmoma/diagnostic imaging , Neurilemmoma/surgery , Retrospective Studies
8.
Exp Cell Res ; 388(2): 111857, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31972221

ABSTRACT

Bone resorption, caused by osteoclasts (OCs), is important to bone homeostasis. The abnormalities of bone resorption may induce a series of diseases, including osteoarthritis, osteoporosis and aseptic peri-implant loosening. The latest research developed,a novel tyrosine and phosphoinositide kinase dual inhibitor, named PP121, inhibited Src in anaplastic thyroid carcinoma cell. However, the therapeutic function of PP121 on abnormal bone resorption is still uncertain. In the present study, we showed that PP121 could potently suppress osteoclast differentiation, osteoclast-specific gene expression and bone resorption via suppressing Src/MAPK (ERK and p38)/Akt-mediated NFATc1 induction in vitro. \It was found that PP121 could suppress the formation of osteoclasts from bone marrow macrophages (BMMs) without causing cytotoxicity, inhibit bone resorption and downregulate the mRNA level of osteoclast-specific markers, including calcitonin receptor (CTR), tartrate resistant acid phosphatase (TRAP), cathepsin K (CTSK), matrix metalloproteinase 3 (MMP3), Cellular oncogene fos (C-Fos) and nuclear factor of activated T-cells cytoplasmic 1 (NFATc1). Consistent with in vitro observation, we found that PP121 greatly ameliorated LPS-induced bone resorption. Our results provide promising evidence of the therapeutic potential of PP121 for osteolytic diseases related to excessive osteoclast-mediated bone resorption.


Subject(s)
Bone Resorption/drug therapy , Cell Differentiation , Lipopolysaccharides/toxicity , Osteoclasts/drug effects , Osteogenesis , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RANK Ligand/metabolism , Animals , Bone Resorption/chemically induced , Bone Resorption/metabolism , Bone Resorption/pathology , Male , Mice , Mice, Inbred C57BL , Osteoclasts/metabolism , RANK Ligand/genetics
9.
J Nanobiotechnology ; 19(1): 157, 2021 May 26.
Article in English | MEDLINE | ID: mdl-34039370

ABSTRACT

BACKGROUND: Smart nanoscale drug delivery systems that target acidic tumor microenvironments (TME) could offer controlled release of drugs and modulate the hypoxic TME to enhance cancer therapy. The majority of previously reported MnO2 nanostructures are nanoparticles, nanosheets, or nanocomposites incorporated with other types of nanoparticles, which may not offer the most effective method for drug loading or for the controlled release of therapeutic payloads. Previous studies have designed MnO2 nanoshells that achieve tumor-specific and enhanced combination therapy for localized advanced cancer. However, the therapeutic effect of MnO2 nanoshells on metastatic cancer is still uncertain. RESULT: Here, intelligent "theranostic" platforms were synthesized based on hollow mesoporous MnO2 (H-MnO2) nanoshells that were loaded with chemotherapy agents docetaxel and cisplatin (TP) to form H-MnO2-PEG/TP nanoshells, which were designed to alleviate tumor hypoxia, attenuate angiogenesis, trigger the dissolution of Mn2+, and synergize the efficacy of first-class anticancer chemotherapy. The obtained H-MnO2-PEG/TP nanoshells decomposed in the acidic TME, releasing the loaded drugs (TP) and simultaneously attenuated tumor hypoxia and hypoxia-inducible factor-1α (HIF-1α) expression by inducing endogenous tumor hydrogen peroxide (H2O2) decomposition. In vitro experiments showed that compared with the control group, the proliferation, colony formation and migration ability of CAL27 and SCC7 cells were significantly reduced in H-MnO2-PEG/TP group, while cell apoptosis was enhanced, and the expression of hypoxia-inducible factor-1α(HIF-1α) was down-regulated. In vivo experiments showed that tumor to normal organ uptake ratio (T/N ratio) of mice in H-MnO2-PEG/TP group was significantly higher than that in TP group alone (without the nanoparticle), and tumor growth was partially delayed. In the H-MnO2-PEG/TP treatment group, HE staining showed that most of the tumor cells were severely damaged, and TUNEL assay showed cell apoptosis was up-regulated. He staining of renal and liver sections showed no obvious fibrosis, necrosis or hypertrophy, indicating good biosafety. Fluorescence staining showed that HIF-1α expression was decreased, suggesting that the accumulation of MnO2 in the tumor caused the decomposition of H2O2 into O2 and alleviated the hypoxia of the tumor. CONCLUSION: In conclusion, a remarkable in vivo and in vitro synergistic therapeutic effect is achieved through the combination of TP chemotherapy, which simultaneously triggered a series of antiangiogenic and oxidative antitumor reactions.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Drug Therapy/methods , Hypoxia/drug therapy , Manganese Compounds/chemistry , Mouth Neoplasms/drug therapy , Nanoshells/chemistry , Squamous Cell Carcinoma of Head and Neck/drug therapy , Tumor Microenvironment/drug effects , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Delivery Systems , Head and Neck Neoplasms/drug therapy , Humans , Hydrogen Peroxide/metabolism , Hydrogen-Ion Concentration , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Oxides/chemistry , Theranostic Nanomedicine/methods , Tumor Hypoxia/drug effects
10.
J Oral Pathol Med ; 49(8): 787-795, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32449223

ABSTRACT

BACKGROUND: To investigate the prognostic value of lymph node ratio (LNR), as well as the correlation with docetaxel, cisplatin, and 5-FU (TPF) induction chemotherapy, in patients with locally advanced oral squamous cell carcinoma (OSCC). METHODS: Two-hundred and forty-five patients from a phase 3 trial involving TPF induction chemotherapy in stage III/IVA OSCC patients (NCT01542931) were enrolled in this study between 2008 and 2010. The clinical and pathological data were collected and analyzed. The cutoff value for LNR was calculated on the receiver operating characteristic (ROC) curve. Univariate and multivariate Cox regression models, and Kaplan-Meier method were used for survival analysis. RESULTS: According to the ROC curve, the cutoff value for LNR was 7.6%. With a median follow-up period of 80 months, the OSCC patients with high-risk LNR (> 7.6%), or positive extranodal extension (ENE) had significantly worse clinical outcomes than patients with low-risk LNR (≤7.6%) or negative ENE. Multivariate analysis on pathological covariates showed that only high-risk LNR was an independent negative predictive factor for survival (P < .05). The cutoff value of LNR of 7.6% was also verified with the similar results using an open TCGA database, high-risk LNR indicating worse overall survival (P < .001) and disease-free survival (P < .001). CONCLUSION: Oral squamous cell carcinoma patients with high-risk LNR have a worse clinical outcome than patients with low-risk LNR. High-risk LNR is an independent negative predictive factor for clinical outcome in patients with locally advanced OSCC.


Subject(s)
Carcinoma, Squamous Cell , Mouth Neoplasms , Neoplasm Staging , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/surgery , Follow-Up Studies , Humans , Lymph Node Excision , Lymph Node Ratio , Lymph Nodes , Lymphatic Metastasis , Mouth Neoplasms/drug therapy , Prognosis , Retrospective Studies
11.
Exp Cell Res ; 382(1): 111470, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31211955

ABSTRACT

Bone resorption, initiated by osteoclasts (OCs), plays an essential role in bone homeostasis. The abnormalities of bone resorption may induce a series of diseases, including osteoarthritis, osteoporosis and aseptic peri-implant loosening. Nirogacestat (PF-03084014, PF), a novel gamma-secretase inhibitor, has been used in phase II clinical trial for treatment of desmoid tumor. However, whether it has the therapeutic effect on abnormal bone resorption remains to be evaluated. In this study, we investigated the role of PF in the regulation of receptor activator of nuclear factor-kB ligand (RANKL)-induced osteoclastogenesis in vitro, and the lipopolysaccharide (LPS)-induced bone resorption in vivo. It was found that PF could suppress the formation of osteoclasts from bone marrow macrophages (BMMs) without causing cytotoxicity, inhibit bone resorption and downregulate the mRNA level of osteoclast-specific markers, including calcitonin receptor (CTR), tartrate resistant acid phosphatase (TRAP), cathepsin K (CTSK), dendritic cell-specific transmembrane protein (Dc-stamp), Atp6v0d2 (V-ATPase d2) and nuclear factor of activated T-cells cytoplasmic 1 (NFATc1). Furthermore, Notch2 signaling, as well as RANKL-induced AKT signaling was significantly inhibited in BMMs. Consistent with in vitro observation, we found that PF greatly ameliorated LPS-induced bone resorption. Taken together, our study demonstrated that PF has a great potential to be used in management of osteolytic diseases.


Subject(s)
Bone Resorption/drug therapy , Macrophages/drug effects , Osteoclasts/drug effects , Osteogenesis/drug effects , Tetrahydronaphthalenes/therapeutic use , Valine/analogs & derivatives , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Bone Resorption/chemically induced , Cells, Cultured , Drug Evaluation, Preclinical , Lipopolysaccharides/toxicity , Macrophage Colony-Stimulating Factor/pharmacology , Male , Mice , Mice, Inbred C57BL , Osteolysis/chemically induced , Osteolysis/drug therapy , RANK Ligand/pharmacology , Recombinant Proteins/pharmacology , Tetrahydronaphthalenes/pharmacology , Valine/pharmacology , Valine/therapeutic use
12.
J Cell Physiol ; 234(7): 10432-10444, 2019 07.
Article in English | MEDLINE | ID: mdl-30652303

ABSTRACT

Chronic periodontitis (CP) is one of the most common oral diseases, which is characterized by the loss of connective tissue and alveolar bone in adults. AZD8835, a novel dual phosphoinositide-3-kinase (PI3K) inhibitor, is currently in phase 1 clinical evaluation to treat breast cancer. However, whether AZD8835 has any effect on teeth and alveolar bone health remains unclear. In the current study, we aimed to investigate the potential effect of AZD8835 in treating CP in vitro and in vivo. We found that AZD8835 could inhibit osteoclast differentiation, bone resorption, and downregulate the expression of osteoclast marker genes, such as tartrate-resistant acid phosphatase (Trap), cathepsin K (Ctsk), V-ATPase d2 (Atp6v0d2), and calcitonin receptor (Ctr). In addition, AZD8835 suppressed osteoclastogenesis by inhibiting receptor activator of nuclear factor kappa B ligand (RANKL)-induced PI3K/protein kinase B (AKT), extracellular signal-regulated kinase, and nuclear factor-κB signaling in BMMs. In vivo, AZD8835 greatly ameliorated alveolar bone (ABL) loss in rats with CP. Meanwhile, histological examination showed fewer osteoclasts in the treatment group. In conclusion, these results indicated that AZD8835 is a promising agent to reduce ABL in CP.


Subject(s)
Alveolar Bone Loss/drug therapy , Osteogenesis/drug effects , Oxadiazoles/pharmacology , Periodontitis/drug therapy , Piperidines/pharmacology , Alveolar Bone Loss/metabolism , Animals , Biomarkers/metabolism , Bone Resorption/drug therapy , Bone Resorption/metabolism , Cathepsin K/metabolism , Cell Line , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Periodontitis/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RANK Ligand/metabolism , Rats , Signal Transduction/drug effects
13.
J Cell Physiol ; 234(11): 20944-20956, 2019 11.
Article in English | MEDLINE | ID: mdl-31020651

ABSTRACT

A series of osteolytic bone diseases are usually related to excessive bone resorption and osteoclast formation. Thus, agents or drugs which can target osteoclast development and attenuate bone loss are potentially considerable in preventing and treating of bone lytic diseases. In recent years, many studies have reported that Notch signaling has substantial impacts on the process of osteoclast differentiation, maturation, and bone destruction. In the present study, we showed that LY411575, a γ-secretase inhibitor, could potently suppress osteoclast differentiation, osteoclast-specific gene expression, and bone resorption via suppressing Notch/HES1/MAPK (ERK and p38)/Akt-mediated NFATc1 induction in vitro. Consistent with in vitro results, LY411575 exhibited protective effects in lipopolysaccharides-induced calvarial bone destruction in vivo. Collectively, these results indicate that LY411575 may have therapeutic potential in the treatment of osteoclast-mediated osteolytic bone diseases.


Subject(s)
Alanine/analogs & derivatives , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Azepines/pharmacology , Enzyme Inhibitors/pharmacology , Osteogenesis/drug effects , Osteolysis/chemically induced , Osteolysis/pathology , Skull/pathology , Actins/metabolism , Alanine/pharmacology , Amyloid Precursor Protein Secretases/metabolism , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Resorption/complications , Bone Resorption/genetics , Bone Resorption/pathology , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Fusion , Cell Movement/drug effects , Cell Movement/genetics , Gene Expression Regulation/drug effects , Lipopolysaccharides , Male , Mice, Inbred C57BL , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , Osteogenesis/genetics , Osteolysis/complications , Osteolysis/genetics , Podosomes/drug effects , Podosomes/metabolism , Protective Agents/pharmacology , RANK Ligand/pharmacology , Signal Transduction/drug effects
16.
Inorg Chem ; 57(7): 3833-3839, 2018 Apr 02.
Article in English | MEDLINE | ID: mdl-29561148

ABSTRACT

The integration of terpyridyl and tricarboxylate functionality in a novel ligand allows concerted 3:1 stoichiometric assembly of size-and charge-complementary Zn2+/Tb3+ ions into a water-stable 3D luminescent framework (CTGU-8) capable of highly selective, sensitive, and recyclable of nitrofurans.

18.
J Pathol ; 236(4): 467-78, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25866254

ABSTRACT

Semaphorin-3F (SEMA3F), an axonal repulsant in nerve development, has been shown to inhibit the progression of human colorectal cancer (CRC); however, the underlying mechanism remains elusive. In this study we found a negative correlation between the levels of SEMA3F and CXCR4 in CRC specimens from 85 patients, confirmed by bioinformatics analysis of gene expression in 229 CRC samples from the Cancer Genome Atlas. SEMA3F(high) /CXCR4(low) patients showed the lowest frequency of lymph node and distant metastasis and the longest survival. Mechanistically, SEMA3F inhibited the invasion and metastasis of CRC cells through PI3K-AKT-dependent down-regulation of the ASCL2-CXCR4 axis. Specifically, ASCL2 enhanced the invasion and metastasis of CRC cells in vitro and expression of ASCL2 correlated with distant metastasis, tumour size and poor overall survival in CRC patients. Treatment of CRC cells with the CXCR4 antagonist AMD3100 attenuated SEMA3F knockdown-induced invasion and metastasis of CRC cells in vitro and in vivo. Our study thus demonstrates that SEMA3F functions as a suppressor of CRC metastasis via down-regulating the ASCL2-CXCR4 axis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Movement , Colorectal Neoplasms/enzymology , Liver Neoplasms/enzymology , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, CXCR4/metabolism , Signal Transduction , Animals , Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Movement/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Computational Biology , Female , Gene Expression Regulation, Neoplastic , Genomics , HCT116 Cells , Humans , Kaplan-Meier Estimate , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Lymphatic Metastasis , Male , Membrane Proteins/genetics , Mice, Nude , Middle Aged , Neoplasm Staging , Nerve Tissue Proteins/genetics , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/genetics , Retrospective Studies , Signal Transduction/drug effects , Time Factors , Transfection , Tumor Burden
19.
J Craniofac Surg ; 25(5): e467-70, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25148639

ABSTRACT

PURPOSE: The aims of this study were to present an uncommon intracranial germinoma manifesting as skull base extension and analyze its clinical characteristics to give valuable insight into such uncommon radiologic variant. METHODS: This is a clinical study of a 15-year-old girl with intracranial germinoma manifesting as skull base extension. Clinical characteristics, magnetic resonance imaging scan observations, pathologic findings, and flow of the treatment procedure were presented and analyzed. RESULTS: She had a 5-month history of diuresis and diplopia. magnetic resonance imaging observation displayed a neoplasm located in the right-side central skull base and suprasellar area with wide extension into the cavernous sinus, intraorbital region, ethmoidal sinus, sphenoid sinus, and pituitary fossa. After administration of contrast medium, strong and heterogeneous enhancement of the mass was observed, with a dural tail sign along the right cerebellar tentorial. Right pterional approach was performed, and intraoperative histologic examination suspected the diagnosis of germinoma; partial resection was achieved, and postoperative radiotherapy was administered. Cranial nerve palsy improved greatly 6 months postoperatively. CONCLUSIONS: Although highly unusual, germinoma should be included in the differential diagnosis of all masses with extension along the midline region of skull base, especially when it happens in young female patients.


Subject(s)
Brain Neoplasms/diagnosis , Germinoma/diagnosis , Skull Base Neoplasms/diagnosis , Adolescent , Diagnosis, Differential , Female , Humans , Magnetic Resonance Imaging
20.
Pharmazie ; 69(10): 752-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25985565

ABSTRACT

Glioma stem cells (GSCs) have been proven to play key roles in tumorigenesis, metastasis and recurrence. Although dihydroartemisinin (DHA), a derivative of the antimalaria drug artemisinin, has been shown to have anti-cancer activity, it is still unclear whether DHA affects GSCs. This study investigated the effects of DHA on the growth and apoptosis of GSCs, as well as the possible molecular mechanism involved in these processes. GSCs were enriched using a non-adhesive culture system with serum-free neural stem cell medium. Their stemness characteristics were identified by assessment of tumor sphere formation, mRNA expression analysis, and immunofluorescence staining of stem cell markers (CD133, SOX2, and nestin). We found that DHA not only inhibited proliferation, which was determined with the cell counting kit-8 (CCK8) assay, but also induced apoptosis of GSCs, as evaluated with the annexin-V/PI flowcytometric assay. Interestingly, DHA treatment also induced a concentration-dependent cell cycle arrest in the G1 phase according to the cell cycle assay. To reveal the underlying mechanisms, we detected the expression levels of p-Akt and Cleaved Caspase-3. The data showed that Cleaved Caspase-3 increased significantly in a dose-dependent manner (p < 0.05) after the GSCs sphere cells were treated with 20, 40, and 80 µM of DHA for 24 h, which correlated with significantly decreased expression levels of p-Akt (p < 0.05). These data indicate that DHA selectively inhibits proliferation and induces apoptosis of GSCs through the down-regulation of Akt phosphorylation, which is followed by Caspase-3 activation, and these findings offer a new approach for treating gliomas.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Artemisinins/pharmacology , Caspase 3/metabolism , Enzyme Activators/pharmacology , Neoplastic Stem Cells/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , G1 Phase/drug effects , Glioma/drug therapy , Mice , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL