Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Cancers (Basel) ; 16(12)2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38927910

ABSTRACT

The therapeutic potential of targeting the ß-catenin/CBP interaction has been demonstrated in a variety of preclinical tumor models with a small molecule inhibitor, ICG-001, characterized as a ß-catenin/CBP antagonist. Despite the high binding specificity of ICG-001 for the N-terminus of CBP, this ß-catenin/CBP antagonist exhibits pleiotropic effects. Our recent studies found global changes in three-dimensional (3D) chromatin architecture in response to disruption of the ß-catenin/CBP interaction in pancreatic cancer cells. However, an understanding of how the functional crosstalk between the antagonist and the ß-catenin/CBP interaction affects changes in 3D chromatin architecture and, thereby, gene expression and downstream effects remains to be elucidated. Here, we perform Hi-C analyses on canonical and patient-derived pancreatic cancer cells before and after treatment with ICG-001. In addition to global alteration of 3D chromatin domains, we unexpectedly identify insulin signaling genes enriched in the altered chromatin domains. We further demonstrate that the chromatin loops associated with insulin signaling genes are significantly weakened after ICG-001 treatment. We finally elicit the deletion of a looping of IRS1-a key insulin signaling gene-significantly impeding pancreatic cancer cell growth, indicating that looping-mediated insulin signaling might act as an oncogenic pathway to promote pancreatic cancer progression. Our work shows that targeting aberrant insulin chromatin looping in pancreatic cancer might provide a therapeutic benefit.

2.
Biomater Sci ; 12(2): 361-374, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-37982147

ABSTRACT

The eyes have a complicated microenvironment with many clearance mechanisms, making it challenging for effective drug delivery to the targeted areas of the eyes. Substrate transport mediated by active transporters is an important way to change drug metabolism in the ocular microenvironment. We designed multifunctional, dual-adaptive nanomicelles (GSCQ@NTB) which could overcome multiple physiological barriers by acting on both the efflux transporter and influx transporter to achieve deep delivery of the P-gp substrate in the cornea. Specifically, an effective "triple" antiangiogenic agent, nintedanib (NTB), was loaded into the biocompatible micelles. The expression of the efflux transporter was reversed by grafting quercetin. The peptide (glycylsarcosine, GS) was modified to target the influx transporter "Peptide Transporter-1" (PepT-1). Quercetin (QRT) and nintedanib (NTB) were transported to the cornea cooperatively, achieving long retention on the ocular surface and high compatibility. In a New Zealand rabbit model, within 8 hours after local administration, GSCQ@NTB was enriched in corneal stromal neovascularization and effectively inhibited the progress of neovascularization. Its effectiveness is slightly better than that in the first-line clinical application of steroids. In this study, we introduce the preparation of a dual adaptive nano-micelle system, which may provide an effective non-invasive treatment for corneal neovascularization.


Subject(s)
Cornea , Quercetin , Animals , Rabbits , Cornea/metabolism , Drug Delivery Systems , Micelles , Biological Transport, Active
3.
bioRxiv ; 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-38013997

ABSTRACT

The therapeutic potential of targeting the ß-catenin/CBP interaction has been demonstrated in a variety of preclinical tumor models with a small molecule inhibitor, ICG-001, characterized as a ß-catenin/CBP antagonist. Despite the high binding specificity of ICG-001 for the N-terminus of CBP, this ß-catenin/CBP antagonist exhibits pleiotropic effects. Our recent studies found global changes in three-dimensional (3D) chromatin architecture in response to disruption of the ß-catenin/CBP interaction in pancreatic cancer cells. However, an understanding of the functional crosstalk between antagonizing the ß-catenin/CBP interaction effect changes in 3D chromatin architecture and thereby gene expression and downstream effects remains to be elucidated. Here we perform Hi-C analyses on canonical and patient-derived pancreatic cancer cells before and after the treatment with ICG-001. In addition to global alteration of 3D chromatin domains, we unexpectedly identify insulin signaling genes enriched in the altered chromatin domains. We further demonstrate the chromatin loops associated with insulin signaling genes are significantly weakened after ICG-001 treatment. We finally elicit the deletion of a looping of IRS1, a key insulin signaling gene, significantly impede pancreatic cancer cell growth, indicating that looping-mediated insulin signaling might act as an oncogenic pathway to promote pancreatic cancer progression. Our work shows that targeting aberrant insulin chromatin looping in pancreatic cancer might provide a therapeutic benefit.

4.
ACS Nano ; 17(24): 24883-24900, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-37883579

ABSTRACT

Surgical resection is the first-line therapy for breast cancer. However, residual tumor cells and the highly immunosuppressive tumor microenvironment (TME) continue to have a serious impact on tumor recurrence and metastasis postresection. Implantation of an in situ hydrogel system postresection has shown to be an effective treatment with great clinical potential. Herein, an injectable zwitterionic hydrogel system was developed for local drug delivery with enhanced immune activation and prevention of tumor recurrence. Driven by electrostatic interactions, poly(sulfobetaine methacrylate) (PSBMA) self-assembles into a hydrogel in saline, achieving low protein adsorption and tunable biodegradability. The chemotherapy drug doxorubicin (DOX) was loaded into copper peroxide nanoparticles (CuO2/DOX), which were coated with macrophage membranes to form tumor-targeting nanoparticles (M/CuO2/DOX). Next, M/CuO2/DOX and the stimulator of interferon genes (STING) agonist 2',3'-cGAMP were coloaded into PSBMA hydrogel (Gel@M/CuO2/DOX/STING). The hydrophilic STING agonist was first released by diffusion from hydrogel to activate the STING pathway and upregulate interferon (IFN) signaling related genes, remodeling the immunosuppressive TME. Then, M/CuO2/DOX targeted the residual tumor cells, combining with DOX-induced DNA damage, immunogenic tumor cell death, and copper death. Hence, this work combines chemodynamic therapy with STING pathway activation in TME, encouraging residual tumor cell death, promoting the maturation of dendritic cells, enhancing tumor-specific CD8+ T cell infiltration, and preventing postoperative recurrence and metastasis.


Subject(s)
Hydrogels , Nanoparticles , Humans , Neoplasm Recurrence, Local/drug therapy , Copper , Neoplasm, Residual/drug therapy , Tumor Microenvironment , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Interferons , Cell Line, Tumor
5.
Biomater Sci ; 11(18): 6325-6341, 2023 Sep 12.
Article in English | MEDLINE | ID: mdl-37555273

ABSTRACT

Triple-negative breast cancer (TNBC) is one of the most aggressive cancers with an immunosuppressive microenvironment, and achieving a satisfactory effect from monotherapies, such as chemotherapy, photodynamic therapy (PDT) or immunotherapy, remains difficult. To solve this puzzle, a deepening synergistic therapy strategy of DNA damage and immunogenic cell death (ICD) stimuli was proposed. We engineered a doxorubicin (DOX) and 4-(hydroxymethyl) phenylboronic acid pinacol ester (PBAP) prodrug polymer, and encapsulated chlorin e6 (Ce6) to obtain the hyaluronidase (HAase) and H2O2 dual-sensitive responsive nanoparticles (Ce6/HDP NPs). The NPs displayed efficient intratumoral accumulation and cellular internalization properties due to the active targeting of the hyaluronic acid (HA). The dual DNA damage of the chemotherapy and ROS production directly caused tumor cell apoptosis. The strong ICD stimuli, which were induced by ROS production and GSH depletion, generated an amplified immunogenicity to activate tumor immunotherapy in vivo. In this manner, the NPs could significantly inhibit primary tumor, abscopal tumor, pulmonary metastasis and recurrent tumor in a subcutaneous 4T1 tumor model, with effective biosafety. This study has provided a promising deepening synergistic therapy strategy against TNBC.


Subject(s)
Nanoparticles , Photochemotherapy , Porphyrins , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Hydrogen Peroxide , Immunogenic Cell Death , Porphyrins/pharmacology , Cell Line, Tumor , Photosensitizing Agents , Tumor Microenvironment
6.
ACS Nano ; 17(12): 11537-11556, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37272777

ABSTRACT

Ferroptosis activation has been considered a mighty weapon for cancer treatment, and growing attention is being paid to reinforcing tumor cells' sensitivity to ferroptosis. However, the existence of certain ferroptosis resistance mechanisms, especially the abnormal metabolism of tumor cells, has long been underestimated. We propose an enhanced ferroptosis-activating pattern via regulating tumor cells' glycometabolism and construct a nanoplatform named PMVL, which is composed of lonidamine (LND)-loaded tannic acid coordinated vanadium oxides with the camouflage of PD-L1 inhibiting peptide-modified tumor cell membrane. This work reveals that the mixed valence of vanadium (VIV and VV) in PMVL triggers ferroptosis due to the self-cyclic valence alteration of V, the process of which generates •OH for lipid peroxide accumulation (VIV → VV) and depletes glutathione (GSH) for glutathione peroxidase (GPX4) deactivation (VV → VIV). Notably, LND strengthens ferroptosis by dual suppression of glycolysis (decreasing ATP supply) and the pentose phosphate pathway (decreasing NADPH production), causing anabatic GSH consumption. Besides, the inhibited glycolysis generates less intracellular lactic acid and alleviates the acidity of tumor microenvironment, preventing immunosuppressive M2 macrophage polarization. In vitro and in vivo data demonstrate the glycometabolism-intervention-enhanced ferroptosis and boosted immunity activation, potentially providing opportunities and possibilities for synergetic cancer therapy.


Subject(s)
Neoplasms , Vanadium , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Cell Death , Glutathione Peroxidase/metabolism , Glucose , Cell Line, Tumor , Tumor Microenvironment
7.
Int J Bioprint ; 9(2): 668, 2023.
Article in English | MEDLINE | ID: mdl-37065667

ABSTRACT

Porous hydroxyapatite (HA) scaffolds prepared by three-dimensional (3D) printing have wide application prospects owing to personalized structural design and excellent biocompatibility. However, the lack of antimicrobial properties limits its widespread use. In this study, a porous ceramic scaffold was fabricated by digital light processing (DLP) method. The multilayer chitosan/alginate composite coatings prepared by layer-by-layer method were applied to scaffolds and Zn2+ was doped into coatings in the form of ion crosslinking. The chemical composition and morphology of coatings were characterized by scanning electron microscope (SEM) and X-ray photoelectron spectroscopy (XPS). Energy dispersive spectroscopy (EDS) analysis demonstrated that Zn2+ was uniformly distributed in the coating. Besides, the compressive strength of coated scaffolds (11.52 ± 0.3 MPa) was slightly improved compared with that of bare scaffolds (10.42 ± 0.56 MPa). The result of soaking experiment indicated that coated scaffolds exhibited delayed degradation. In vitro experiments demonstrated that within the limits of concentration, a higher Zn content in the coating has a stronger capacity to promote cell adhesion, proliferation and differentiation. Although excessive release of Zn2+ led to cytotoxicity, it presented a stronger antibacterial effect against Escherichia coli (99.4%) and Staphylococcus aureus (93%).

8.
J Drug Target ; 31(4): 335-353, 2023 04.
Article in English | MEDLINE | ID: mdl-36543743

ABSTRACT

Extracellular vesicles (EVs) could be produced by most cells and play an important role in disease development. As a subtype of EVs, exosomes exhibit suitable size, rich surface markers and diverse contents, making them more appealing as potential drug carriers. Compared with traditional synthetic nanoparticles, exosomes possess superior biocompatibility and much lower immunogenicity. This work reviewed the most up-to-date research progress of exosomes as carriers for nucleic acids, proteins and small molecule drugs for cancer and inflammation management. The drug loading strategies and potential cellular uptake behaviour of exosomes are highlighted, trying to provide reference for future exosome design and application.


Exosomes are secreted by a variety of cells and play an important role in the process of inter-cell communication.This paper provides a comprehensive review focussing on the up-to-date applications of exosomes as carriers of nucleic acids, proteins and small molecule drugs for cancer and inflammation management.This paper briefly introduces the basic properties of exosomes, from definition, biogenesis to cellular uptake manners.Various strategies to enable exosomes to efficiently load cargoes are highlighted.Problems to be solved when using exosomes to deliver drugs are discussed.


Subject(s)
Exosomes , Extracellular Vesicles , Neoplasms , Humans , Drug Carriers/metabolism , Exosomes/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Drug Delivery Systems
9.
Biomaterials ; 290: 121832, 2022 11.
Article in English | MEDLINE | ID: mdl-36228518

ABSTRACT

Photodynamic therapy (PDT) induces apoptosis of cancer cells by generating cytotoxic reactive oxygen species, the therapeutic effect of which, however, is impeded by intrinsic/inducible apoptosis-resistant mechanisms in cancer cells and hypoxia of tumor microenvironment (TME); also, PDT-induced anti-tumor immunity activation is insufficient. To deal with these obstacles, a novel biomimetic nanoplatform is fabricated for the precise delivery of photosensitizer chlorin e6 (Ce6), hemin and PEP20 (CD47 inhibitory peptide), integrating oxygen-boosted PDT, ferroptosis activation and CD47-SIRPα blockade. Hemin's catalase-mimetic activity alleviates TME hypoxia and enhances PDT. The nanoplatform activates ferroptosis via both classical (down-regulating glutathione peroxidase 4 pathway) and non-classical (inducing Fe2+ overload) modes. Besides the role of hemin in consuming glutathione and up-regulating heme oxygenase-1 expression, interestingly, we observe that Ce6 enhance ferroptosis activation via both classical and non-classical modes. The anti-cancer immunity is reinforced by combining PEP20-mediated CD47-SIRPα blockade and PDT-mediated T cell activation, efficiently suppressing primary tumor growth and metastasis. PEP20 has been revealed for the first time to sensitize ferroptosis by down-regulating system Xc-. This work sheds new light on the mechanisms of PDT-ferroptosis activation interplay and bridges immunotherapy and ferroptosis activation, laying the theoretical foundation for novel combinational modes of cancer treatment.


Subject(s)
Chlorophyllides , Ferroptosis , Photochemotherapy , Porphyrins , Humans , CD47 Antigen , Tumor Microenvironment , Oxygen/pharmacology , Biomimetics , Hemin/pharmacology , Chlorophyllides/pharmacology , Cell Line, Tumor , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Phototherapy , Hypoxia/drug therapy
11.
Signal Transduct Target Ther ; 7(1): 130, 2022 04 25.
Article in English | MEDLINE | ID: mdl-35462576

ABSTRACT

Oral squamous cell carcinoma (OSCC) is the most common type of oral malignancy, and metastasis accounts for the poor prognosis of OSCC. Autophagy is considered to facilitate OSCC development by mitigating various cellular stresses; nevertheless, the mechanisms of autophagy in OSCC cell proliferation and metastasis remain unknown. In our study, high-sensitivity label-free quantitative proteomics analysis revealed nuclear protein 1 (NUPR1) as the most significantly upregulated protein in formalin-fixed paraffin-embedded tumour samples derived from OSCC patients with or without lymphatic metastasis. Moreover, NUPR1 is aberrantly expressed in the OSCC tissues and predicts low overall survival rates for OSCC patients. Notably, based on tandem mass tag-based quantitative proteomic analysis between stable NUPR1 knockdown OSCC cells and scrambled control OSCC cells, we confirmed that NUPR1 maintained autophagic flux and lysosomal functions by directly increasing transcription factor E3 (TFE3) activity, which promoted OSCC cell proliferation and metastasis in vitro and in vivo. Collectively, our data revealed that the NUPR1-TFE3 axis is a critical regulator of the autophagic machinery in OSCC progression, and this study may provide a potential therapeutic target for the treatment of OSCC.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Basic Helix-Loop-Helix Transcription Factors , Head and Neck Neoplasms , Neoplasm Proteins , Squamous Cell Carcinoma of Head and Neck , Autophagy/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation/genetics , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Neoplasm Proteins/genetics , Nuclear Proteins , Proteomics , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology
12.
J Mech Behav Biomed Mater ; 121: 104642, 2021 09.
Article in English | MEDLINE | ID: mdl-34174680

ABSTRACT

Calcium silicate is a common implant material with excellent mechanical strength and good biological activity. In recent years, the addition of strengthening materials to calcium silicate has been proven to promote bone tissue regeneration, but its degradation properties require further improvements. In this paper, calcium silicate was used as the matrix, and 10 wt% hydroxyapatite and 10 wt% strontium phosphate were added to im prove the biological activity of the scaffold. The effect of adding different amounts of calcium sulfate dihydrate (CaSO4·2H2O) on the degradation of the scaffold was explored. A porous ceramic scaffold was prepared by digital light processing (DLP) technology, and its performance was evaluated. Cell experiments showed that the addition of calcium sulfate improved cell proliferation and differentiation. Simulated body fluid (SBF) immersion tests showed that small amounts of apatite deposits appeared on the fourth day, larger deposits appeared on the 14th day, and degradation occurred on the surface after 28 days of immersion. Mechanical tests showed that the addition of 5 wt% CaSO4·2H2O improved the compressibility of the composite. After soaking in SBF for 14 days, it retained its compressive strength (11.8 MPa), which meets the requirements of cancellous bone, demonstrating its potential application value for bone repair.


Subject(s)
Calcium Compounds , Silicates , Ceramics , Printing, Three-Dimensional , Tissue Scaffolds
13.
J Cell Mol Med ; 25(4): 2262-2273, 2021 02.
Article in English | MEDLINE | ID: mdl-33345447

ABSTRACT

Oral submucosal fibrosis (OSF) is one of the pre-cancerous lesions of oral squamous cell carcinoma (OSCC). Its malignant rate is increasing, but the mechanism of malignancy is not clear. We previously have elucidated the long non-coding RNA (lncRNA) expression profile during OSF progression at the genome-wide level. However, the role of lncRNA ADAMTS9-AS2 in OSF progression via extracellular communication remains unclear. lncRNA ADAMTS9-AS2 is down-regulated in OSCC tissues compared with OSF and normal mucous tissues. Low ADAMTS9-AS2 expression is associated with poor overall survival. ADAMTS9-AS2 is frequently methylated in OSCC tissues, but not in normal oral mucous and OSF tissues, suggesting tumour-specific methylation. Functional studies reveal that exosomal ADAMTS9-AS2 suppresses OSCC cell growth, migration and invasion in vitro. Mechanistically, exosomal ADAMTS9-AS2 inhibits AKT signalling pathway and regulates epithelial-mesenchymal transition markers. Through profiling miRNA expression profile regulated by exosomal ADAMTS9-AS2, significantly enriched pathways include metabolic pathway, PI3K-Akt signalling pathway and pathways in cancer, indicating that exosomal ADAMTS9-AS2 exerts its functions through interacting with miRNAs during OSF progression. Thus, our findings highlight the crucial role of ADAMTS9-AS2 in the cell microenvironment during OSF carcinogenesis, which is expected to become a marker for early diagnosis of OSCC.


Subject(s)
Exosomes/metabolism , Oral Submucous Fibrosis/genetics , Oral Submucous Fibrosis/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Long Noncoding/genetics , Signal Transduction , Cell Communication , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Exosomes/ultrastructure , Gene Expression Profiling , Gene Expression Regulation , Humans , Methylation , MicroRNAs/genetics , Oral Submucous Fibrosis/pathology , Prognosis , RNA, Long Noncoding/metabolism
14.
J Drug Target ; 29(1): 12-28, 2021 01.
Article in English | MEDLINE | ID: mdl-32698651

ABSTRACT

At present, malignant tumours have become one of the most serious diseases that endanger human health. According to a survey on causes of death in Chinese population in early 1990s, the malignant tumours were the second leading cause of death. In the treatment of tumours, the ideal situation is that drugs should target and accumulate at tumour sites and destroy tumour cells specifically, without affecting normal cells and stem cells with regenerative capacity. This requires drugs to be specifically transported to the target organs, tissues, cells, and even specific organelles, like mitochondria, nuclei, lysosomes, endoplasmic reticulum (ER), and Golgi apparatus (GA). The nano drug delivery system can not only protect drugs from degradation but also facilitate functional modification and targeted drug delivery to the tumour site. This article mainly reviews the targeting of nano drug delivery systems to tumour cytoplasmic matrix, nucleus, mitochondria, ER, and lysosomes. Organelle-specific drug delivery system will be a major mean of targeting drug delivery with lower toxicity, less dosage and higher drug concentration in tumour cells.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems/trends , Nanoparticles/administration & dosage , Neoplastic Cells, Circulating/drug effects , Organelles/drug effects , Animals , Antineoplastic Agents/metabolism , Drug Delivery Systems/methods , Humans , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Organelles/metabolism , Organelles/pathology , Tumor Microenvironment/drug effects , Tumor Microenvironment/physiology
15.
Biochim Biophys Acta Gene Regul Mech ; 1863(11): 194631, 2020 11.
Article in English | MEDLINE | ID: mdl-32956836

ABSTRACT

BACKGROUND: Recent studies suggested that crosstalk between ERα and EGFR/HER2 pathways plays a critical role in mediating endocrine therapy resistance. Several inhibitors targeting EGFR/HER2 signaling, including FDA-approved lapatinib and gefitinib as well as a novel dual tyrosine kinase inhibitor (TKI) sapitinib, showed greater therapeutic efficacies. However, how 3D chromatin landscape responds to the inhibition of EGFR/HER2 pathway remains to be elucidated. METHODS: In this study, we conducted in situ Hi-C and RNA-seq in two ERα+ breast cancer cell systems, 1) parental MCF7 cells and its associated tamoxifen-resistant MCF7TR cells; and 2) parental T47D cells and its associated tamoxifen-resistant T47DTR cells, before and after the treatment of sapitinib. RESULTS: We identified differential responses in topologically associated domains (TADs), looping genes and expressed genes. Interestingly, we found that many differential TADs and looping genes are reversible after sapitinib treatment, indicating that EGFR/HER2 signaling may play a role in reshaping and rewiring the high order genome organization. We further examined and recapitulated the reversible looping genes in 3D spheroids of breast cancer cells, demonstrating that 3D cell culture spheroid of breast cancer cells could be a potential preclinical breast cancer model for studying 3D chromatin regulation. CONCLUSIONS: Our study has provided significant insights into our understanding of 3D genomic landscape changes in response to EGFR/HER2 Inhibition in endocrine-resistant breast cancer cells. Our data provides a rich resource for further evaluating chromatin structural responses to EGFR/HER2 targeted therapies in endocrine-resistant breast cancer cells. Our analyses suggest that these alterations of chromatin structures and transcriptional programs may provide new avenues for intervention or designing of patient selection for targeted endocrine treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Genomics , Protein Kinase Inhibitors/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms , Cell Line, Tumor , Computational Biology/methods , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Female , Gene Expression Profiling , Gene Ontology , Genomics/methods , Humans , Signal Transduction/drug effects
16.
Oral Oncol ; 110: 104866, 2020 11.
Article in English | MEDLINE | ID: mdl-32623343

ABSTRACT

OBJECTIVES: Reconstruction of the total tongue after cancer resection remains one of the challenges in head and neck surgery. Inadequate reconstruction after subtotal or total glossectomy defects leads to poor quality of life. The aim of this study was to explore an economical, practical and effective flap design for functional tongue reconstruction. MATERIAL AND METHODS: Sixty patients were randomly divided into two groups, namely, a "Sushi roll" technique group (30 patients) and a conventional surgery group (30 patients). Then, the patients underwent total or subtotal tongue reconstruction. Swallowing function, speech intelligibility, cosmetic results, and quality of life were assessed with the appropriate scales. Outcomes were analysed, and a p-value <0.05 was considered significant. RESULTS: The perioperative recovery of the "Sushi roll" group was superior to that of the conventional group. Relative to patients in the conventional group, patients in the "Sushi roll" group showed significantly improved speech intelligibility (p = 0.025), cosmetic results (p < 0.001) and swallowing function (p < 0.001). CONCLUSION: The innovative "Sushi roll" anterolateral thigh myocutaneous flap approach for total tongue reconstruction creates a free neotongue tip with adequate volume and protuberance and causes minimal damage to the donor site, producing acceptable swallowing function and speech intelligibility.


Subject(s)
Myocutaneous Flap/surgery , Plastic Surgery Procedures/methods , Thigh/surgery , Tongue Neoplasms/surgery , Tongue/pathology , Female , Humans , Male , Middle Aged
17.
J Cancer ; 10(19): 4540-4551, 2019.
Article in English | MEDLINE | ID: mdl-31528218

ABSTRACT

As a master regulator of embryonic morphogenesis, homeodomain-containing gene 10 (HOXC10) has been found to promote progression of human cancers and indicate poor survival outcome. Therefore, we concentrate on elucidating the role of HOXC10 in progression of oral squamous cell carcinoma (OSCC). In our study, the expression of HOXC10 was significantly increased in human OSCC samples and was significantly correlated with TNM stage and lymph node metastasis. Upregulation of HOXC10 indicated a poor overall survival of OSCC patients according to the Kaplan-Meier survival curves. Furthermore, HOXC10-knockdown dramatically suppressed migration, invasion, and expression of N-Cadherin, Vimentin and Snail, as well as increased E-cadherin level both in vivo and in vitro. Bioinformatics and cellular study further confirmed that HOXC10 may promote invasion and migration of OSCC cells by regulating the WNT/epithelial-mesenchymal transition (EMT) signaling pathway. These findings suggest that HOXC10 plays a pivotal role in the metastasis of OSCC and highlight its usefulness as a potential prognostic marker or therapeutic target in human OSCC.

18.
J Oncol ; 2019: 6835176, 2019.
Article in English | MEDLINE | ID: mdl-31467541

ABSTRACT

Oral submucous fibrosis (OSF) as one of the premalignant disorders endures a series of histopathological stages to invasive oral squamous cell carcinoma (OSCC) eventually. However, the role of long non-coding RNA (lncRNA) expression in OSF malignant progression still remains poorly understood. Through RNA-sequencing normal mucous, OSF and OSCC tissues, we found 687 lncRNA transcripts significantly and differentially expressed during OSF progression, including 231 upregulated lncRNAs and 456 downregulated lncRNAs, indicating that lncRNAs are involved in the regulation of different stages of OSF development. Further functional enrichment analysis showed these differentially expressed lncRNAs participated in inflammation signaling, Wnt signaling, angiogenesis, CCKR signaling, integrin signaling, PDGF signaling, p53 signaling, and EGF receptor (EGFR) signaling pathways, which contribute to inflammatory and fibroelastic pathogenetic changes of OSF and further malignant progression. Five novel lncRNAs were differentially expressed during OSF progression with varied expression levels, indicating the importance of these lncRNAs in OSF malignant development. Moreover, some lncRNAs have been previously identified to be associated with OSCC pathogenesis, including HCG22, RP11-397A16.1, LINC00271, CTD-3179P9.1, and ZNF667-AS1. Thus, our study firstly comprehensively elucidated lncRNAs expression profile of malignant procession from OSF premalignant lesion to OSCC, which will enlighten our understanding of the importance of lncRNA involved in OSF malignant development.

19.
Toxicol Lett ; 314: 142-152, 2019 Oct 10.
Article in English | MEDLINE | ID: mdl-31319114

ABSTRACT

Cadmium (Cd), an established carcinogen, is a risk factor for oral squamous cell carcinoma (OSCC). Macroautophagy/autophagy is proposed to play a pivotal role in Cd-mediated carcinogenic activity. However, the mechanisms underlying Cd-induced autophagy are poorly understood. In the present study, a CAL27 OSCC cell line exposed to 10-6 M Cd for 8 weeks was used as a model system. Repeated Cd exposure induced significant migration and invasion of CAL27 cells. Furthermore, we showed that Cd increased the autophagic flux in CAL27 cells, as evidenced by the upregulation of LC3-II and the downregulation of P62/SQSTM1. The genetic blocking of autophagy inhibited Cd-induced migration and invasion, indicating a carcinogenic role of autophagy in Cd-treated CAL27 cells. Cd-induced NUPR1 expression, which contributes to lysosomal biogenesis and expression of autophagy-related gene, was found to mechanistically initiate autophagy in CAL27 cells. Of note, NUPR1 shRNA abolished Cd-induced autophagy both in vitro and in vivo. We also found that Cd triggered the generation of MDA in a xenograft tumour model and that N-acetyl-l-cysteine, a reactive oxygen species (ROS) scavenger, abrogated the effects of Cd on NUPR1-dependent autophagy in vivo. Taken together, these results demonstrate that ROS-dependent NUPR1-mediated autophagy plays an important role in repeated Cd exposure -induced cell growth, migration and invasion in OSCC cells.


Subject(s)
Acetylcysteine/pharmacology , Antioxidants/pharmacology , Autophagy/drug effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cadmium Chloride/toxicity , Cell Movement/drug effects , Mouth Neoplasms/drug therapy , Neoplasm Proteins/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Squamous Cell Carcinoma of Head and Neck/drug therapy , Animals , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Mice, Nude , Microtubule-Associated Proteins/metabolism , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Sequestosome-1 Protein/metabolism , Signal Transduction , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
20.
BMC Cancer ; 18(1): 911, 2018 Sep 21.
Article in English | MEDLINE | ID: mdl-30241505

ABSTRACT

Traditional therapeutics have encountered a bottleneck caused by diagnosis delay and subjective and unreliable assessment. Biomarkers can overcome this bottleneck and guide us toward personalized precision medicine for oral squamous cell carcinoma. To achieve this, it is important to efficiently and accurately screen out specific biomarkers from among the huge number of molecules. Progress in omics-based high-throughput technology has laid a solid foundation for biomarker discovery. With credible and systemic biomarker models, more precise and personalized diagnosis and assessment would be achieved and patients would be more likely to be cured and have a higher quality of life. However, this is not straightforward owing to the complexity of molecules involved in tumorigenesis. In this context, there is a need to focus on tumor heterogeneity and homogeneity, which are discussed in detail. In this review, we aim to provide an understanding of biomarker discovery and application for precision medicine of oral squamous cell carcinoma, and have a strong belief that biomarker will pave the road toward future precision medicine.


Subject(s)
Biomarkers, Tumor , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/metabolism , Mouth Neoplasms/etiology , Mouth Neoplasms/metabolism , Animals , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/therapy , Early Detection of Cancer , Epigenomics/methods , Genetic Heterogeneity , Genomics/methods , High-Throughput Screening Assays , Humans , Mouth Neoplasms/diagnosis , Mouth Neoplasms/therapy , Precision Medicine/methods , Proteomics/methods , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...