Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Discov Oncol ; 15(1): 82, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38512482

ABSTRACT

OBJECTIVE: Synovial Sarcoma (SS), a highly malignant mesenchymal neoplasm, typically carries a grim prognosis for patients presenting with high-grade or metastatic disease. Although Anlotinib, a new agent for treating soft tissue sarcomas, holds promise, its underlying mechanism remains incompletely understood. This investigation aims to delineate Anlotinib's anticancer effectiveness and potential mechanistic underpinnings in patients suffering from advanced, refractory SS. MATERIALS AND METHODS: Employing microarray assay, we examined the potential downstream targets of Anlotinib in SS therapy. A shRNA-based high-content screening was performed to identify candidate genes with the greatest influence on SW982 cell proliferation. The knockdown efficacy of selected genes within SW982 cells was confirmed using RT-qPCR as well as western blot analysis. To assess the effect of putative downstream elimination of genes with synovial sarcoma cells, cell proliferation, and apoptotic assays were carried out. Gene chip microarray as well as bioinformatics techniques were utilized to scrutinize potential signaling networks associated with the candidate downstream gene. RESULTS: QPCR verified high expression of FAM83D in SW982 cells, shRNA was designed to silence FAM83D by lentivirus transfection, apoptosis assay, and cell cycle arrest showing that FAM83D downregulation augments apoptosis in SW982 cells and arrests cell cycle progression in the S stage. Inhibition of FAM83D expression upregulated STAT1 while downregulated BIRC5, MCM2, and CDK1 genes in vitro. CONCLUSIONS: This experimental study identified FAM83D as a critical regulator that contributes to the proliferation and progression of SS, suggesting that FAM83D-regulated signaling pathway may serve as a prospective target in SS management.

2.
Oncol Lett ; 12(3): 1806-1810, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27588127

ABSTRACT

The aim of the present study was to assess the efficacy and safety of topical timolol maleate combined with oral propranolol for parotid infantile hemangiomas. Between October 2012 and April 2014, propranolol was administered orally at a dose of 1.0-1.5 mg/kg/day to 22 infants with proliferating hemangiomas in the Department of Oral and Maxillofacial Surgery (Hospital of Stomatology, China Medical University, Shenyang, Liaoning, China). A small amount of 0.5% timolol maleate eye drop solution was topically applied with medical cotton swabs to the area of the lesion twice a day, every 12 h. The study group consisted of 9 males and 13 females, aged 2-9 months, with a median age of 4.7 months. The lesions were all located in the parotid region, and measured between 3.5×4×0.5 and 7×8×3 cm in volume. The planned duration of therapy was 6-8 months, or the two drugs were stopped when complete regression of the lesions was obtained. The therapeutic outcomes and safety were assessed by the change in the size and color of the tumor, and the presence of adverse effects throughout the course of treatment. The mean duration of therapy was 21.1 weeks and ranged from 3 to 8 months. Of the 22 patients, 16 demonstrated an excellent response, 6 showed a good response and 2 displayed a moderate response. No major collateral effects were observed. Overall, oral propranolol combined with topical timolol maleate may be used as the first-line therapeutic choice in the treatment of infantile parotid mixed hemangioma.

3.
Int J Mol Med ; 38(2): 367-80, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27352815

ABSTRACT

The role of transforming growth factor-ß1 (TGF-ß1) in normal human fracture healing has been previously demonstrated. The objective of the present study was to examine the biocompatibility of TGF-ß1-silk fibroin-chitosan (TGF-ß1-SF-CS) three-dimensional (3D) scaffolds in order to construct an ideal scaffold for bone tissue engineering. We added TGF-ß1 directly to the SF-CS scaffold to construct a 3D scaffold for the first time, to the best of our knowledge, and performed evaluations to determine whether it may have potential applications as a growth factor delivery device. Bone marrow-derived mesenchymal stem cells (BMSCs) were seeded on the TGF-ß1-SF-CS scaffolds and the silk fibroin-chitosan (SF-CS) scaffolds. On the TGF-ß1­SF-CS and the SF-CS scaffolds, the cell adhesion rate increased in a time­dependent manner. Using a Cell Counting Kit-8 (CCK-8) assay and analyzing the alkaline phosphatase (ALP) expression proved that TGF-ß1 significantly enhanced the growth and proliferation of BMSCs on the SF-CS scaffolds in a time-dependent manner. To examine the in vivo biocompatibility and osteogenesis of the TGF-ß1­SF-CS scaffolds, the TGF-ß1-SF-CS scaffolds and the SF-CS scaffolds were implanted in rabbit mandibles and studied histologically and microradiographically. The 3D computed tomography (CT) scan and histological examinations of the samples showed that the TGF-ß1-SF-CS scaffolds exhibited good biocompatibility and extensive osteoconductivity with the host bone after 8 weeks. Moreover, the introduction of TGF-ß1 to the SF-CS scaffolds markedly enhanced the efficiency of new bone formation, and this was confirmed using bone mineral density (BMD) and biomechanical evaluation, particularly at 8 weeks after implantation. We demonstrated that the TGF-ß1­SF-CS scaffolds possessed as good biocompatibility and osteogenesis as the hybrid ones. Taken together, these findings indicate that the TGF-ß1-SF-CS scaffolds fulfilled the basic requirements of bone tissue engineering, and have the potential to be applied in orthopedic, reconstructive and maxillofacial surgery. Thus, TGF-ß1-SF-CS composite scaffolds represent a promising, novel type of scaffold for use in bone tissue engineering.


Subject(s)
Chitosan/pharmacology , Fibroins/pharmacology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Transforming Growth Factor beta1/pharmacology , Absorption, Physicochemical , Alkaline Phosphatase/metabolism , Animals , Biomechanical Phenomena/drug effects , Bone Density/drug effects , Bone Regeneration/drug effects , Cell Adhesion/drug effects , Cell Count , Cell Differentiation/drug effects , Cell Shape/drug effects , Cells, Cultured , Female , Imaging, Three-Dimensional , Male , Mandible/diagnostic imaging , Mandible/drug effects , Mandible/pathology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/ultrastructure , Organ Size/drug effects , Osteogenesis/drug effects , Porosity , Rabbits , Tomography, X-Ray Computed
4.
J Oral Maxillofac Surg ; 74(11): 2193-2199, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27235180

ABSTRACT

PURPOSE: Periorbital infantile hemangiomas (IHs) require early intervention because they have the potential risk of causing visual disturbances. In recent years, propranolol has shown promise in the effective management of periocular and periorbital IHs. The objective of our study was to assess the clinical outcomes, efficacy, and safety of propranolol in the management of infants with high-risk periorbital IHs. PATIENTS AND METHODS: This retrospective study was conducted at the Stomatological Hospital affiliated with China Medical University. The medical records of infants with periorbital hemangiomas who were treated with systemic propranolol at a dose of 1.0 to 1.5 mg/kg per day between January 2014 and June 2015 were reviewed. We excluded infants who did not qualify for propranolol treatment and infants who received previous therapy or other treatments. The records were reviewed for treatment response, adverse events during treatment, length of treatment, and recurrences. Treatment response was classified using a 4-point scale system based on reduction in volume as poor (<25%), moderate (25 to 50%), good (50 to 75%), or excellent (>75 to 100%) and change in color, as well as surface texture, by a panel of 3 plastic surgeons using 2-dimensional photographs, clinical examination, and Doppler ultrasonography measurements taken before and after treatment. RESULTS: Of 38 infants with periorbital hemangiomas, 26 were treated with systemic propranolol at a dose of 1.0 to 1.5 mg/kg administered once daily. A total of 11 male and 15 female infants with a mean age of 5.2 months (range, 2 to 12 months) were treated. The mean length of treatment was 22 weeks (range, 4 to 41 weeks). Adverse events of diarrhea (n = 3) and sleep changes (n = 1) were encountered during treatment in 4 patients. The overall treatment response was scored as excellent in 17 patients, good in 7, moderate in 2, and poor in 0. No patients required discontinuation of treatment because of adverse events, and there were no cases of recurrence or tumor regrowth noted during the mean follow-up period of 6.5 months (range, 3 to 10 months). CONCLUSIONS: Oral propranolol at a dose of 1.0 to 1.5 mg/kg per day (age ≤3 months, 1.0 mg/kg; age >3 months, 1.5 mg/kg) was effective and well tolerated for the management of 26 Chinese infants with high-risk periorbital IHs. Early intervention should be considered to reduce risk of visual impairment and improve esthetic outcomes.


Subject(s)
Adrenergic beta-Antagonists/therapeutic use , Hemangioma/drug therapy , Propranolol/therapeutic use , Skin Neoplasms/drug therapy , Administration, Oral , Drug Administration Schedule , Eye , Female , Follow-Up Studies , Humans , Infant , Male , Retrospective Studies , Treatment Outcome
5.
J Oral Maxillofac Surg ; 74(8): 1623-9, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27055227

ABSTRACT

PURPOSE: Infantile hemangiomas (IHs) are the most common benign tumors affecting infants, and most IHs are self-limiting. However, there are cases that require specific treatment. Propranolol is now widely used to treat severe IHs. Several studies have shown the efficacy and limited side effects associated with propranolol as the first-line treatment for IHs. There are a limited number of publications describing the role of propranolol in treating IHs beyond the proliferative phase (>12 months). The purpose of this study was to evaluate the effects and safety of oral high-dose (2.0 mg/kg per day) propranolol for IHs beyond the proliferative phase (>12 months). PATIENTS AND METHODS: This study enrolled patients with IHs who accepted systemic propranolol treatment from the Department of Oral and Maxillofacial Surgery, Stomatological Hospital Affiliated China Medical University. This is a single-center retrospective study conducted from April 2011 to July 2015. All children who were older than 12 months were eligible for the study. Digital photographs taken before and after treatment were analyzed by a panel of 3 plastic surgeons. The esthetic results were evaluated using a 4-point scale and ranked as poor, moderate, good, or excellent. The patient follow-up visits were scheduled monthly, and changes in the size, texture, and color of the lesions were recorded. The adverse effects after medication were evaluated and managed accordingly. RESULTS: We collected data on 31 eligible patients. The 31 patients had 32 hemangiomas (1 female patient had 2 lesions) and were treated with systemic propranolol at a high dose of 2 mg/kg per day. The mean age at the initiation of propranolol therapy was 18.4 months (range, 12 to 48 months), and the mean treatment duration was 10.1 months (range, 8 to 16 months). The treatment responses for the 32 hemangiomas included 17 excellent responses (53.1%), 8 good responses (25%), and 7 moderate responses (21.9%). There were no severe side effects encountered and recurrence was observed in 3 patients during the treatment and follow-up course. CONCLUSIONS: Oral propranolol, 2 mg/kg per day, is a safe and effective treatment for IHs beyond the proliferative phase (>12 months of age) in the Chinese population.


Subject(s)
Head and Neck Neoplasms/drug therapy , Hemangioma/drug therapy , Propranolol/therapeutic use , Vasodilator Agents/therapeutic use , Administration, Oral , Child, Preschool , Female , Humans , Infant , Male , Propranolol/administration & dosage , Retrospective Studies , Vasodilator Agents/administration & dosage
6.
J Craniofac Surg ; 27(2): 509-15, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26890455

ABSTRACT

The objective of the study was to discuss the biocompatibility of the vascular endothelial growth factor-silk fibroin-chitosan (VEGF-SF-CS) scaffolds. To offer an ideal scaffold for bone tissue engineering, the author added vascular endothelial growth factor (VEGF) into silk fibroin-chitosan (SF-CS) scaffold directly to reconstruct a three-dimensional scaffold for the first time, SF-CS scaffold was loaded with VEGF and evaluated as a growth factor-delivery device. Human fetal osteoblast cell was seeded on the VEGF-SF-CS scaffolds and SF-CS scaffolds. On VEGF-SF-CS and SF-CS scaffolds, the cell adhesion rate was increased as time went on. Scanning electron microscopy: the cells grew actively and had normal multiple fissions, granular and filamentous substrates could be seen around the cells, and cell microfilaments were closely connected with the scaffolds. The cells could not only show the attached growth on surfaces of the scaffolds, but also extend into the scaffolds. Cell Counting Kit-8 and alkaline phosphatase analysis proved that the VEGF could significantly promote human fetal osteoblast1.19 cells growth and proliferation in the SF-CS scaffolds, but the enhancement of osteoblasts cell proliferation and activity by VEGF was dependent on time.


Subject(s)
Chitosan/chemistry , Fibroins/chemistry , Osteogenesis/physiology , Silk/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Vascular Endothelial Growth Factor A/chemistry , Actin Cytoskeleton/physiology , Actin Cytoskeleton/ultrastructure , Alkaline Phosphatase/analysis , Biocompatible Materials/chemistry , Cell Adhesion/physiology , Cell Count , Cell Culture Techniques , Cell Proliferation , Cell Shape/physiology , Cells, Cultured , Humans , Materials Testing , Microscopy, Electron, Scanning , Osteoblasts/physiology , Osteoblasts/ultrastructure , Surface Properties
7.
Mol Med Rep ; 13(1): 181-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26530112

ABSTRACT

The aim of the present study was to examine the biocompatibility of transforming growth factor-ß1-silk fibroin-chitosan (TGF-ß1-SF-CS) scaffolds. In order to provide an ideal scaffold for use in bone tissue engineering, TGF-ß1 was introduced into the SF­CS scaffold in order to reconstruct a three dimensional scaffold, following which hFOB1.19 osteoblast cells were seeded onto TGF­ß1­SF­CS and SF­CS scaffolds. On the TGF­ß1­SF­CS and SF­CS scaffolds, the cell adhesion rate increased in a time­dependent manner. Scanning electron microscopy revealed that the cells grew actively and exhibited normal morphological features with multiple fissions, and granular and filamentous substrates were observed surrounding the cells. In addition, the cell microfilaments were closely connected with the scaffolds. The cells exhibited attached growth on the surfaces of the scaffolds, however, the growth also extended into the scaffolds. Cell Counting Kit­8 and ALP analyses revealed that TGF­ß1 significantly promoted the growth and proliferation of the hFOB1.19 osteoblast cells in the SF­CS scaffolds, and the enhancement of osteoblast cell proliferation and activity by TGF­ß1 occurred in a time­dependent manner. The TGF-ß1-SF-CS composite material may offer potential as an ideal scaffold material for bone tissue engineering.


Subject(s)
Cell Culture Techniques/methods , Chitosan/pharmacology , Fetus/cytology , Fibroins/pharmacology , Osteoblasts/cytology , Tissue Scaffolds/chemistry , Transforming Growth Factor beta1/pharmacology , Alkaline Phosphatase/metabolism , Cell Adhesion/drug effects , Cell Count , Cell Proliferation/drug effects , Cell Shape/drug effects , Humans , Osteoblasts/drug effects
8.
Dent Mater J ; 34(4): 475-84, 2015.
Article in English | MEDLINE | ID: mdl-26235712

ABSTRACT

The objective of this study was to discuss the construction method, characterization, and biocompatibility of three-dimensional silk fibroin-chitosan (SF-CS) scaffolds which met the requirements of bone tissue engineering scaffolds. Silk fibroin (SF) and chitosan (CS) were mixed at different ratios -3 to 7, 5 to 5, and 7 to 3- to fabricate the composite materials. To find out the optimum mixing ratio of SF and CS, parameters such as pore size, porosity, water absorption, and the mechanical properties were evaluated. Osteoblast cells hFOB1.19 were seeded on SF-CS scaffolds and pure CS scaffolds for the first time. Cell adhesion rate, cell proliferation, and cell activity were evaluated, and cell growth and formation of mineralized nodules were observed. Results showed that SF-CS scaffolds are a suitable candidate for bone tissue engineering.


Subject(s)
Chitosan/chemistry , Fibroins/chemistry , Osteoblasts/physiology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Biocompatible Materials , Cell Adhesion , Cell Line , Humans , Materials Testing , Microscopy, Electron, Scanning , Surface Properties , Water
9.
J Craniofac Surg ; 26(2): 438-40, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25692890

ABSTRACT

The aim of our study was to assess the efficacy and safety of oral propranolol for the treatment of parotid infantile hemangiomas. Between October 2009 and January 2013, propranolol was given orally to 30 infants with proliferating hemangiomas at a dose of 1.0 to 1.5 mg/kg per day in our department. The patients included 12 male infants and 18 female infants, aged between 2 and 13 months, with a median of 5.9 months. The lesions were located in the parotid region and measured from 1.5 cm × 2 cm × 0.5 cm to 6 cm × 8 cm × 3 cm in volume. Oral propranolol was administered once daily for a mean duration of 22.7 weeks (range, 14-32 wk). Follow-up times were from 1 to 10 months (median, 6.4 mo). Changes in the color and size of the tumor were recorded using hemisphere measurements and digital photographs. The treatment results were scored according to a 4-point scale. Overall response was graded scale 4 (excellent) in 18 patients, scale 3 (good) in 11 patients, scale 2 (moderate) in 1 patient, and scale 1 (poor) in none. No major collateral effects and rebounds were observed in any of the patients. Oral propranolol was a well-tolerated and effective treatment with mild adverse effects for parotid infantile hemangiomas.


Subject(s)
Adrenergic beta-Antagonists/therapeutic use , Hemangioma/drug therapy , Parotid Neoplasms/drug therapy , Propranolol/therapeutic use , Administration, Oral , Adrenergic beta-Antagonists/administration & dosage , Color , Female , Follow-Up Studies , Hemangioma/pathology , Humans , Infant , Male , Parotid Neoplasms/pathology , Photography/methods , Propranolol/administration & dosage , Remission Induction , Safety , Treatment Outcome , Ultrasonography, Doppler
SELECTION OF CITATIONS
SEARCH DETAIL
...