Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
J Nanobiotechnology ; 21(1): 201, 2023 Jun 26.
Article in English | MEDLINE | ID: mdl-37365598

ABSTRACT

Malignant bone tumors result in high rates of disability and death and are difficult to treat in terms of killing tumors and repairing bone defects. Compared with other hyperthermia strategies, magnetic hyperthermia has become an effective therapy for treating malignant bone tumors due to its lack of depth limitations. However, tumor cells express heat shock protein (HSP) to resist hyperthermia, which reduces its curative effect. Competitive ATP consumption can reduce HSP production; fortunately, the basic principle of starvation therapy by glucose oxidase (GOx) is consuming glucose to control ATP production, thereby restricting HSP generation. We developed a triple-functional magnetic gel (Fe3O4/GOx/MgCO3@PLGA) as a magnetic bone repair hydrogels (MBRs) with liquid‒solid phase transition capability to drive magneto-thermal effects to simultaneously trigger GOx release and inhibit ATP production, reducing HSP expression and thereby achieving synergistic therapy for osteosarcoma treatment. Moreover, magnetic hyperthermia improves the effect of starvation therapy on the hypoxic microenvironment and achieves a reciprocal strengthening therapeutic effect. We further demonstrated that in situ MBRs injection effectively suppressed tumor growth in 143B osteosarcoma tumor-bearing mice and an in-situ bone tumor model in the rabbit tibial plateau. More importantly, our study also showed that liquid MBRs could effectively match bone defects and accelerate their reconstruction via magnesium ion release and enhanced osteogenic differentiation to augment the regeneration of bone defects caused by bone tumors, which generates fresh insight into malignant bone tumor treatment and the acceleration of bone defect repair.


Subject(s)
Bone Neoplasms , Hyperthermia, Induced , Osteosarcoma , Mice , Animals , Rabbits , Osteogenesis , Bone Neoplasms/therapy , Bone Neoplasms/metabolism , Osteosarcoma/therapy , Osteosarcoma/metabolism , Bone Regeneration , Heat-Shock Proteins/metabolism , Magnetic Phenomena , Adenosine Triphosphate , Cell Line, Tumor , Tumor Microenvironment
2.
Onco Targets Ther ; 11: 3989-4000, 2018.
Article in English | MEDLINE | ID: mdl-30022842

ABSTRACT

BACKGROUND: Tubeimoside-1 (TBMS1), a triterpenoid saponin extracted from traditional Chinese medicine tubeimoside, exerts a cytotoxic effect on several human cancer cell lines. However, no study has focused on whether TBMS1 works on oral squamous cell carcinoma (OSCC). MATERIALS AND METHODS: We treated OSCC cells with TBMS1 to detect the effect and relevant molecular basis of TBMS1 for the first time. We chose two oral cancer cell lines, CAL27 and SCC15, for this study. First, the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenylte-trazolium bromide assay and cell proliferation 5'-bromo-2'-deoxyuridine assay were carried out to detect cell growth. Second, colony formation assay was performed to assess clonogenesis capacity. Next apoptosis was analyzed by flow cytometry. Subsequently, wound healing and transwell assays were applied to explore cell migration. Finally, Western blot was further performed to examine corresponding proteins' expression change. RESULTS: Our data showed that TBMS1 significantly suppressed proliferation of OSCC cells in a dose- and time-dependent manner and it inhibited migration of OSCC cells as well. After treatment with TBMS1, OSCC cells underwent cell apoptosis. Furthermore, Western blot demonstrated that TBMS1 downregulated apoptosis-associated proteins such as PARP, p-ERK1/2, Bcl-2, caspase-3, caspase-7 and caspase-8 and upregulated cleaved PARP, cleaved caspase-3 and cleaved caspase-9. It could also reduce expression of c-Myc and MMP-7. Meanwhile, TBMS1 did not change the total ERK1/2 expression. CONCLUSION: These results revealed that TBMS1 might be a potential chemotherapeutic drug for the management of OSCC.

3.
J Immunol Res ; 2017: 9786972, 2017.
Article in English | MEDLINE | ID: mdl-28951880

ABSTRACT

A derivative formula, DGBX, which is composed of three herbs (Radix astragali, Radix Angelicae sinensis, and Coptis chinensis Franch), is derived from a famous Chinese herbal formula, Danggui Buxue Tang (DBT) (Radix astragali and Radix Angelicae sinensis). We aimed to investigate the effects of DGBX on the regulation of the balance between proliferation and apoptosis of hematopoietic stem cells (HSCs) due to the aberrant immune response in a mouse model of aplastic anemia (AA). Cyclosporine (CsA), an immunosuppressor, was used as the positive control. Our results indicated that DGBX could downregulate the production of IFNγ in bone marrow cells by interfering with the binding between SLAM and SAP and the expressions of Fyn and T-bet. This herbal formula can also inhibit the activation of Fas-mediated apoptosis, interferon regulatory factor-1-induced JAK/Stat, and eukaryotic initiation factor 2 signaling pathways and thereby induce proliferation and attenuate apoptosis of HSCs. In conclusion, DGBX can relieve the immune-mediated destruction of HSCs, repair hematopoietic failure, and recover the hematopoietic function of HSCs in hematogenesis. Therefore, DGBX can be used in traditional medicine against AA as a complementary and alternative immunosuppressive therapeutic formula.


Subject(s)
Anemia, Aplastic/therapy , Complex Mixtures/therapeutic use , Drugs, Chinese Herbal/therapeutic use , Hematopoietic Stem Cells/pathology , Anemia, Aplastic/immunology , Animals , Apoptosis/drug effects , Cells, Cultured , Chromatography, High Pressure Liquid , Complex Mixtures/analysis , Disease Models, Animal , Female , Hematopoietic Stem Cells/drug effects , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred DBA
4.
PLoS One ; 12(7): e0180417, 2017.
Article in English | MEDLINE | ID: mdl-28683082

ABSTRACT

Angelicae Sinensis, Radix Astragali and Rhizoma Coptidis are all herbs of modified Danggui Buxue Tang (DGBX) and are extensively applied herbs in traditional Chinese medicine for the treatment of anemia and inflammation. In this study, immune-induced AA mice were used as an animal model, and the immunosuppressive agent, Ciclosporin A (CsA), was used as a positive control. Multiple pro-inflammatory cytokines were examined by bead-based multiplex flow cytometry. The T-cell subsets were assessed using a fluorescence-activated cell sorter (FACS). Western blot analysis was used to estimate the protein expression levels of specific transcription factors for T helper cells (Th1, Th2 and Th17) and key molecules of the Janus-activated kinase (Jak)/signal transducer and activator of transcription (Stat3) signaling pathway. DGBX treatment could significantly increase the production of whole blood cells in peripheral blood (PB); inhibit the expansion of Th1 and Th17 cells; increase the differentiation of Th2 and Tregs cells; regulate the expression levels of T-bet, GATA-3, RORγ and proinflammatory cytokines; and decrease the expression levels of key molecules in the Jak/Stat signaling pathway. These results indicate that DGBX can regulate the differentiation of T lymphocytes, resulting in immunosuppressive and hematogenic functions on AA mice. DGBX might be a good candidate for inclusion in a randomized study for AA with more data on the possible side effects and doses used in humans. Ultimately, it may be used for applications of traditional medicine against AA in modern complementary and alternative immunosuppressive therapeutics.


Subject(s)
Anemia, Aplastic/drug therapy , Bone Marrow/drug effects , Drugs, Chinese Herbal/pharmacology , Immunosuppressive Agents/pharmacology , Th1 Cells/drug effects , Th17 Cells/drug effects , Th2 Cells/drug effects , Anemia, Aplastic/genetics , Anemia, Aplastic/immunology , Anemia, Aplastic/pathology , Angelica sinensis/chemistry , Animals , Astragalus propinquus , Bone Marrow/immunology , Bone Marrow/pathology , Cell Differentiation/drug effects , Cyclosporine/pharmacology , Disease Models, Animal , Drugs, Chinese Herbal/chemistry , Female , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/immunology , Humans , Medicine, Chinese Traditional , Mice , Mice, Inbred BALB C , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Ranunculaceae/chemistry , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , Th1 Cells/immunology , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology
5.
Sci Rep ; 7: 41370, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28145487

ABSTRACT

Aditoprim (ADP) has potential use as an antimicrobial agent in animals. However, its pharmacodynamic properties have not been systematically studied yet. In this study, the in vitro antibacterial activities of ADP and its main metabolites were assayed, and the in vivo antibacterial efficacy of ADP for the treatment of swine streptococcosis was evaluated. It was shown that Salmonella and Streptococcus from swine, Escherichia coli and Salmonella from chickens, E. coli, Streptococcus, Mannheimia, Pasteurella from calves, Streptococcus and Mannheimia from sheep, and E. coli, Flavobacterium columnare, Acinetobacter baumannii and Yersinia ruckeri from fishes were highly susceptible to ADP. Haemophilus parasuis from swine, Staphylococcus aureus, Aeromonas punctate, Mycobacterium tuberculosis, Streptococcus agalactiae from fishes, and Klebsiella from calves and sheep showed moderate susceptibility to ADP, whereas E. coli, Actinobacillus pleuropneumonia, Pasteurella, S. aureus, Clostridium perfringens from swine, S. aureus, C. perfringens from chickens, and S. aureus from calves were resistant to ADP. The main metabolites of ADP showed equal activity to that of their parent compound, and the prevention and therapeutic dosages of ADP recommended for swine streptococcosis were 10 and 20~40 mg/kg b.w., respectively. This study firstly showed that ADP had strong antibacterial activity and had potential to be used as a single drug in the treatment of bacterial infectious diseases.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Streptococcal Infections/drug therapy , Swine Diseases/drug therapy , Swine Diseases/microbiology , Trimethoprim/analogs & derivatives , Animals , Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests , Streptococcal Infections/microbiology , Streptococcal Infections/pathology , Swine , Treatment Outcome , Trimethoprim/metabolism , Trimethoprim/pharmacology , Trimethoprim/therapeutic use
6.
J Tradit Chin Med ; 36(5): 606-12, 2016 10.
Article in English | MEDLINE | ID: mdl-29933528

ABSTRACT

OBJECTIVE: To observe the effect of compound Zhebei granules (CZBG) with chemotherapy in the treatment of refractory acute leukemia. METHODS: In this multicenter, double-blind, placebo-controlled clinical trial, we used a central (online) randomization system to assign 235 patients to two treatment groups. A total of 118 patients received chemotherapy combined with CZBG (4 g, twice daily) and 117 patients received chemotherapy plus placebo. The clinical efficacy was evaluated at the end of one chemotherapeutic cycle. RESULTS: In the full analysis set, in which deaths due to disease progression were regarded as inefficacy, the rates of complete remission (CR) and partial remission (CR + PR) were 32.35% and 50.00% , respectively, for the chemotherapy combined with CZBG group, and 23.08% and 35.58%, respectively, for the chemotherapy plus placebo group. There was a statistically significant difference between the two groups according to a χ2 test (P < 0.05). In the per protocol analysis set (PPS), the CR (33.67%), CR+PR (52.04%) response rates for the chemotherapy plus CZBG group were significantly different from the response rates of the control group (CR: 24.24% and CR+PR: 37.37%), respectively (P < 0.05). CONCLUSION: CZBG plus chemotherapy can improve the clinical remission rate of refractory acute leukemia after one just one therapeutic cycle.


Subject(s)
Antineoplastic Agents/administration & dosage , Drugs, Chinese Herbal/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Acute Disease/therapy , Adult , Double-Blind Method , Drug Therapy , Drug Therapy, Combination , Drugs, Chinese Herbal/chemistry , Female , Humans , Male , Middle Aged , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL