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.
Pharmaceutics ; 15(8)2023 Jul 26.
Article in English | MEDLINE | ID: mdl-37631239

ABSTRACT

Hydroxyapatite-gelatin microspheres with cone-like pores were synthesized via the wet-chemical method using ammonium dihydrogen phosphate ((NH4)H2PO4) and calcium nitrate (Ca(NO3)2·4H2O) as a source of calcium and phosphate ions with the addition of gelatin, which proved to be more osteoconductive than commercial products, such as fibrin glue and Osteoset® Bone Graft Substitute. Following the method of the previous study for loading paclitaxel (PTX), a drug entrapment efficiency of around 58% was achieved, which is much lower than that of the doxorubicin (DOX)-loaded one. Since PTX is hydrophobic while DOX is hydrophilic, the order of chitosan processing and addition of the solvent were tuned in this study, finally leading to an increase in drug entrapment efficiency of 94%. Additionally, the release duration of PTX exceeded six months. The MTT assay indicated that the effect of drug release on the suppression of cancer cells reached more than 40% after one week, thereby showcasing PTX's capacity to carry out its medicinal functions without being affected by the loading procedures.

2.
Neurotox Res ; 40(5): 1223-1234, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35829999

ABSTRACT

Adipose tissue-derived mesenchymal stem cells (ADSC) exert neuroprotective and anti-inflammatory effects. ADSCs are considered potential therapeutics for rheumatoid arthritis (RA), an inflammatory, multisystemic autoimmune disease. Epigallocatechin-3-gallate (EGCG), the major polyphenolic compound in green tea, has strong anti-inflammatory and antioxidant properties. This study aimed to investigate whether EGCG has a synergistic effect on the neuroprotective effects of ADSCs to protect the RA-damaged brain. Wistar rats were classified into four groups: sham, RA, RA + ADSCs (1 × 106 cells per rat), and RA + EGCG (10 µM)-pretreated ADSCs. After 2 months of treatment, the brain tissues from the rats were collected and investigated. The brains of RA rats had higher inflammation and apoptosis. ADSC treatment ameliorated these negative effects significantly; however, the neuroprotective abilities of EGCG-pretreated ADSCs were significantly higher than ADSCs. Furthermore, the RA-induced repression of the PI3K/Akt survival pathway was reactivated by EGCG-pretreated ADSCs. Collectively, this study provides evidence that EGCG synergistically enhances the neuroprotective ability of ADSCs to repress the negative effects of RA on the brain. These findings could help develop new therapeutic strategies against RA or other neurodegenerative diseases after clinical validation in the future.


Subject(s)
Arthritis, Rheumatoid , Catechin , Neuroprotective Agents , Animals , Antioxidants/pharmacology , Brain/metabolism , Catechin/analogs & derivatives , Catechin/pharmacology , Collagen/metabolism , Neuroinflammatory Diseases , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Stem Cells/metabolism , Tea
4.
Drug Deliv Transl Res ; 11(4): 1532-1544, 2021 08.
Article in English | MEDLINE | ID: mdl-34125402

ABSTRACT

There is an urgent need for treatments for hydrofluoric acid (HF) burns and their derivative problems that prevent hydrogen ion dissociation and fluoride ion binding to tissues. This study evaluated the ability of chitosan-based hydrogels combined with a buffer solution containing either boric acid or Tris and calcium gluconate (CHS-BA-CG and CHS-Tris-CG) to repair HF burn wounds and prevent wound infections. We assessed calcium release rates and biocompatability and constructed a mouse HF burn model to assess the tissue repair effects of the hydrogels. Finally, we performed disc diffusion tests from burn tissue and quantified the bacterial counts to assess the anti-infection properties of the hydrogels. Calcium was gradually released in the CHS-BA-CG and CHS-Tris-CG groups (73% and 43%, respectively, after 48 h). The cell viabilities at 48 h after HF burn in these groups were significantly higher than those in the phosphate-buffered saline (PBS) and CG-treated groups. Histopathological evaluation showed a clear boundary between the epidermal and dermal layers in both CHS-BA-CG and CHS-Tris-CG-treated groups, indicating their effectiveness in tissue repair. In the disc diffusion test, CHS-BA-CG and CHS-Tris-CG exhibited larger inhibition zones against Acinetobacter baumannii than those for PBS and CG. The bacterial counts on HF burn wounds were significantly lower in the CHS-BA-CG and CHS-Tris-CG-treated groups than those in the PBS and CG-treated groups. The in vitro studies demonstrated the biocompatibility and antimicrobial effects of the CHS-BA-CG and CHS-Tris-CG hydrogels. Both gels also demonstrated tissue repair and anti-infection effects. Thus, chitosan-based hydrogels may be candidates for HF burn therapy.


Subject(s)
Burns, Chemical , Burns , Chitosan , Wound Infection , Animals , Burns/drug therapy , Burns/microbiology , Hydrofluoric Acid , Hydrogels/chemistry , Mice , Wound Infection/drug therapy , Wound Infection/microbiology , Wound Infection/prevention & control
5.
Int J Mol Sci ; 21(9)2020 May 09.
Article in English | MEDLINE | ID: mdl-32397561

ABSTRACT

Although various advancements in radical surgery and neoadjuvant chemotherapy have been developed in treating osteosarcoma (OS), their clinical prognosis remains poor. A synthetic chemical compound, 3-hydroxylflavone, that is reported to regulate ROS production is known to inhibit human bone osteosarcoma cells. However, its role and mechanism in human OS cells remains unclear. In this study, we have determined the potential of 3-Hydroxy-2-phenylchromone (3-HF) against OS using human osteosarcoma (HOS) cells. Our previous studies showed that Zipper sterile-alpha-motif kinase (ZAK), a kinase member of the MAP3K family, was involved in various cellular events such as cell proliferation and cell apoptosis, and encoded two transcriptional variants, ZAKα and ß. In this study, we show that 3-HF induces the expression of ZAK and thereby enhances cellular apoptosis. Using gain of function and loss of function studies, we have demonstrated that ZAK activation by 3-HF in OS cells is confined to a ZAKß form that presumably plays a leading role in triggering ZAKα expression, resulting in an aggravated cancer apoptosis. Our results also validate ZAKß as the predominant form of ZAK to drive the anticancer mechanism in HOS cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bone Neoplasms/pathology , Flavonoids/pharmacology , MAP Kinase Kinase 4/drug effects , MAP Kinase Kinase Kinases/drug effects , Osteosarcoma/pathology , Caspase 3/biosynthesis , Caspase 3/genetics , Cell Line, Tumor , Enzyme Activation/drug effects , Gain of Function Mutation , Humans , Loss of Function Mutation , MAP Kinase Kinase Kinases/genetics , Membrane Potential, Mitochondrial/drug effects , Protein Isoforms/drug effects , Protein Isoforms/genetics , Signal Transduction/drug effects , Up-Regulation/drug effects
6.
Environ Toxicol ; 34(8): 902-911, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31044527

ABSTRACT

Osteosarcoma (OS) is a tumor entity that can cause a large number of cancer-related deaths. Although chemotherapy can decrease proliferation and increase apoptosis of human OS cells, the clinical prognosis remains poor. Fisetin is a flavonol found in fruits and vegetables and is reported to inhibit cell growth in numerous cancers. But the molecular mechanism underlying fisetin in human OS cells is not clear. It is known that sterile-alpha motif and leucine zipper containing kinase (ZAK), a kinase in the MAP3K family, is involved in various cell processes, including proliferation and apoptosis. In our lab, we have demonstrated that overexpression of ZAK can induce apoptosis in human OS cells. In the previous studies, MAP4K, the upstream of MAP3K, can act in parallel to MST1/2 to activate LATS1/2 in the Hippo pathway. Turning on the Hippo pathway can decrease proliferation and otherwise cause cell apoptosis in cancer cells. In this study, we found that fisetin can upregulate ZAK expression to induce the Hippo pathway and mediate the activation of JNK/ERK, the downstream of ZAK, to trigger cell apoptosis via AP-1 dependent manner in human OS cells. These findings reveal a novel molecular mechanism underlying fisetin effect on human OS cells.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Neoplasms/metabolism , Flavonoids/pharmacology , MAP Kinase Signaling System , Osteosarcoma/metabolism , Protein Kinases/metabolism , Apoptosis , Bone Neoplasms/enzymology , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonols , Hippo Signaling Pathway , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinases , Osteosarcoma/enzymology , Osteosarcoma/pathology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transcription Factor AP-1/metabolism , Tumor Suppressor Proteins/metabolism
7.
Cell Biochem Funct ; 36(4): 176-182, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29654619

ABSTRACT

ZAK is a novel mixed lineage kinase-like protein that contains a leucine-zipper and a sterile-alpha motif as a protein-protein interaction domain, and it is located in the cytoplasm. There are 2 alternatively spliced forms of ZAK: ZAKα and ZAKß. Previous studies showed that ZAKα is involved in various cell processes, including cell proliferation, cell differentiation, and cardiac hypertrophy, but the molecular mechanism of ZAKß is not yet known. In a recent study in our laboratory, we found that ZAKß can ameliorate the apoptotic effect induced by ZAKα in H9c2 cells. We further hypothesized that ZAKß could also improve the apoptotic effect induced by ZAKα in human osteosarcoma cells. The results of this study show that ZAKß can induce apoptosis and decrease cell viability similar to the effects of ZAKα. Interestingly, our ZAKα-specific inhibitor assay shows that the expression of ZAKß is highly dependent on ZAKα expression. However, ZAKß expression effectively induces ZAKα expression and results in synergistic enhancement of apoptosis in human osteosarcoma cells. Furthermore, co-immunoprecipitation results revealed that ZAKα can directly interact with ZAKß, and this interaction may contribute to the enhanced apoptotic effects. SIGNIFICANCE OF THE STUDY: ZAK is a mixed lineage kinase involved in cell differentiation, proliferation, and hypertrophic growth. ZAKα isoform of ZAK is associated with tumorigenesis, but the function of ZAKß is not yet known. In H9c2 cells, ZAKß was found to ameliorate the apoptotic effect induced by ZAKα. However, in osteosarcoma cells, ZAKß elevates the apoptotic effect induced by ZAKα. In this study, we show that similar to ZAKα, the ZAKß induces apoptosis and decreases cell viability. Interestingly, the expression of ZAKß is dependent on ZAKα expression, and ZAKß further enhances ZAKα expression and results in synergistic enhancement of apoptosis in osteosarcoma cells.


Subject(s)
Apoptosis/drug effects , Osteosarcoma/metabolism , Protein Kinases/biosynthesis , Antibiotics, Antineoplastic/pharmacology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Drug Synergism , Humans , MAP Kinase Kinase Kinases , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Structure-Activity Relationship , Tumor Cells, Cultured
8.
Environ Toxicol ; 33(2): 191-197, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29105997

ABSTRACT

Human osteosarcoma (OS) is a malignant cancer of the bone. It exhibits a characteristic malignant osteoblastic transformation and produces a diseased osteoid. A previous study demonstrated that doxorubicin (DOX) chemotherapy decreases human OS cell proliferation and might enhance the relative RNA expression of ZAK. However, the impact of ZAKα overexpression on the OS cell proliferation that is inhibited by DOX and the molecular mechanism underlying this effect are not yet known. ZAK is a protein kinase of the MAPKKK family and functions to promote apoptosis. In our study, we found that ZAKα overexpression induced an apoptotic effect in human OS cells. Treatment of human OS cells with DOX enhanced ZAKα expression and decreased cancer cell viability while increasing apoptosis of human OS cells. In the meantime, suppression of ZAKα expression using shRNA and inhibitor D1771 both suppressed the DOX therapeutic effect. These findings reveal a novel molecular mechanism underlying the DOX effect on human OS cells. Taken together, our findings demonstrate that ZAKα enhances the apoptotic effect and decreases cell viability in DOX-treated human OS cells.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Apoptosis/drug effects , Doxorubicin/toxicity , Protein Kinases/metabolism , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , MAP Kinase Kinase Kinases , NF-kappa B/metabolism , Osteosarcoma/metabolism , Osteosarcoma/pathology , Protein Kinases/chemistry , Protein Kinases/genetics , RNA Interference , RNA, Small Interfering/metabolism , bcl-X Protein/metabolism
9.
Cell Biochem Funct ; 34(8): 606-612, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27859413

ABSTRACT

ZAK (sterile alpha motif and leucine zipper containing kinase AZK), a serine/threonine kinase with multiple biochemical functions, has been associated with various cell processes, including cell proliferation, cell differentiation, and cardiac hypertrophy. In our previous reports, we found that the activation of ZAKα signaling was critical for cardiac hypertrophy. In this study, we show that the expression of ZAKα activated apoptosis through both a FAS-dependent pathway and a mitochondria-dependent pathway by subsequently inducing caspase-3. ZAKß, an isoform of ZAKα, is dramatically expressed during cardiac hypertrophy and apoptosis. The interaction between ZAKα and ZAKß was demonstrated here using immunoprecipitation. The results show that ZAKß has the ability to diminish the expression level of ZAKα. These findings reveal an inherent regulatory role of ZAKß to antagonize ZAKα and to subsequently downregulate the cardiac hypertrophy and apoptosis induced by ZAKα.


Subject(s)
Apoptosis , Cardiomegaly/metabolism , Cardiomegaly/pathology , Protein Kinases/metabolism , Animals , Cell Survival , Intracellular Space/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Protein Binding , Rats , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...