Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
BMC Complement Med Ther ; 24(1): 186, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734604

ABSTRACT

BACKGROUND: Cepharanthin® alone or in combination with glucocorticoid (GC) has been used to treat chronic immune thrombocytopenia (ITP) since the 1990s. Cepharanthine (CEP) is one of the main active components of Cepharanthin®. The purpose of this study was to investigate the effects of CEP on GC pharmacodynamics on immune cells and analyse the possible action mechanism of their interactions. METHODS: Peripheral blood mononuclear cells (PBMCs), T lymphocytic leukemia MOLT-4 cells and daunorubicin resistant MOLT-4 cells (MOLT-4/DNR) were used to evaluate the pharmacodynamics and molecular mechanisms. Drug pharmacodynamics was evaluated by WST-8 assay. P-glycoprotein function was examined by rhodamine 123 assay. CD4+CD25+Foxp3+ regulatory T cells and Th1/Th2/Th17 cytokines were detected by flow cytometry. P-glycoprotein expression and GC receptor translocation were examined by Western blot. RESULTS: CEP synergistically increased methylprednisolone (MP) efficacy with the suppressive effect on the cell viability of PBMCs. 0.3 and 1 µM of CEP significantly inhibited P-glycoprotein efflux function of CD4+ cells, CD8+ cells, and lymphocytes (P<0.05). 0.03~3 µM of CEP also inhibited the P-glycoprotein efflux function in MOLT-4/DNR cells in a concentration-dependent manner (P<0.001). However, 0.03~3 µM of CEP did not influence P-glycoprotein expression. 0.03~0.3 µM of CEP significantly increased the GC receptor distribution from the cytoplasm to the nucleus in a concentration-dependent manner in MOLT-4/DNR cells. The combination did not influence the frequency of CD4+, CD4+CD25+ and CD4+CD25+Foxp3+ T cells or the secretion of Th1/Th2/Th17 cytokines from PBMCs. In contrast, CEP alone at 1 µM decreased the percentage of CD4+ T cell significantly (P<0.01). It also inhibited the secretion of IL-6, IL-10, IL-17, TNF-α, and IFN-γ. CONCLUSIONS: CEP synergistically promoted MP pharmacodynamics to decrease the cell viability of the mitogen-activated PBMCs, possibly via inhibiting P-glycoprotein function and potentiating GC receptor translocation. The present study provides new evidence of the therapeutic effect of Cepharanthin® alone or in combination with GC for the management of chronic ITP.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1 , Benzylisoquinolines , Drug Synergism , Leukocytes, Mononuclear , Methylprednisolone , Receptors, Glucocorticoid , Humans , Benzylisoquinolines/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Methylprednisolone/pharmacology , Receptors, Glucocorticoid/metabolism , Benzodioxoles
2.
BMC Pharmacol Toxicol ; 25(1): 13, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38303016

ABSTRACT

BACKGROUND: Methotrexate (MTX) is the cornerstone of rheumatoid arthritis (RA) treatment and is highly effective with low-dose intermittent administration. MTX is occasionally used in combination with non-steroidal anti-inflammatory drugs (NSAIDs) and acetaminophen (APAP)/paracetamol for pain or inflammation control. With MTX treatment, the side effects, such as hepatotoxicity, renal failure, and myelosuppression should be considered. These are also seen with analgesics treatment. METHODS: We used a large spontaneously reported adverse event database (FAERS [JAPIC AERS]) to analyze whether the reporting of adverse events increased upon MTX and analgesic therapy in patients with RA. RESULTS: After identifying RA cases, the crude reporting odds ratios (cRORs) for hepatotoxicity, renal failure, and thrombocytopenia associated with the use of MTX, APAP, or NSAIDs were calculated by disproportionality analysis, which revealed significantly higher cRORs for these events. No analgesics showed consistent positive signals for drug-drug interaction (DDI) with concomitant low-dose MTX analyzed using four algorithms for DDI interaction (the Ω shrinkage measure, additive or multiplicative, and combination risk ratio models). However, in renal failure and thrombocytopenia, loxoprofen (Ω025 = 0.08) and piroxicam (Ω025 = 0.46), and ibuprofen (Ω025 = 0.74) and ketorolac (Ω025 = 3.52), respectively, showed positive signals in the Ω shrinkage measure model, and no consistency was found among adverse events or NSAIDs. CONCLUSIONS: Studies using spontaneous reporting systems have limitations such as reporting bias or lack of patient background; however, the results of our comprehensive analysis support the results of previous clinical or epidemiological studies. This study also demonstrated the usefulness of FAERS for DDI assessment.


Subject(s)
Antirheumatic Agents , Arthritis, Rheumatoid , Chemical and Drug Induced Liver Injury , Renal Insufficiency , Thrombocytopenia , Humans , Methotrexate/adverse effects , Antirheumatic Agents/adverse effects , Acetaminophen/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Analgesics/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/chemically induced , Thrombocytopenia/chemically induced , Thrombocytopenia/drug therapy , Drug Interactions , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/drug therapy , Renal Insufficiency/chemically induced
3.
Front Pharmacol ; 14: 1030832, 2023.
Article in English | MEDLINE | ID: mdl-36909171

ABSTRACT

Methotrexate (MTX) is an essential anti-rheumatic drug used to treat rheumatoid arthritis (RA). Prevention or management of adverse reactions, including interstitial lung disease (ILD), hepatotoxicity, myelosuppression, and infection, remains fundamental for safe MTX therapy. Using the Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS) (JAPIC AERS), we performed disproportionality analyses of adverse events related to MTX use and the impact of concomitant medications. Upon analyzing all reported cases in FAERS between 1997 and 2019, the crude reporting odds ratios (cRORs; 95% confidence intervals) for ILD, hepatotoxicity, myelosuppression, and tuberculosis (TB) in relation to MTX use were 4.00 (3.83-4.17), 1.99 (1.96-2.02), 3.66 (3.58-3.74), and 7.97 (7.65-8.3), respectively. Combining MTX with folic acid (FA) or tumor necrosis factor-alpha inhibitors (TNFis) tended to reduce cRORs for these adverse events (except for TB). Multiple logistic regression analysis in patients with RA was conducted to calculate adjusted reporting odds ratios (aRORs) for age, sex, and MTX treatment patterns (MTX alone and combined with FA and TNFi). Higher age (except for hepatotoxicity) and male sex were significantly associated with adverse events. Combining FA or TNFi with MTX reduced aRORs for MTX-related hepatotoxicity and myelosuppression; in contrast, the effect of FA was not obvious in ILD or TB. Although studies assessing spontaneous reporting systems have limitations such as reporting bias, data from our logistic regression analysis demonstrated that adding FA to MTX-based therapy could help reduce the dose-dependent adverse events of MTX, thereby providing clinical evidence that supports the beneficial effect of FA. This study also demonstrated the usefulness of FAERS in comparing adverse events based on treatment patterns.

4.
Int J Mol Sci ; 23(17)2022 Aug 23.
Article in English | MEDLINE | ID: mdl-36076929

ABSTRACT

Previously, we demonstrated that the proton pump inhibitor, esomeprazole magnesium hydrate (MH), could have potential as a repurposed treatment against preeclampsia, a serious obstetric condition. In this study we investigate the difference in the preclinical effectiveness between 100 µM of esomeprazole MH and its hydration isomer, esomeprazole magnesium trihydrate (MTH). Here, we found that both treatments reduced secretion of sFLT-1 (anti-angiogenic factor) from primary cytotrophoblast, but only esomeprazole MH reduced sFLT-1 secretion from primary human umbilical vein endothelial cells (assessed via ELISA). Both drugs could mitigate expression of the endothelial dysfunction markers, vascular cell adhesion molecule-1 and endothelin-1 (via qPCR). Neither esomeprazole MH nor MTH quenched cytotrophoblast reactive oxygen species production in response to sodium azide (ROS assay). Finally, using wire myography, we demonstrated that both compounds were able to induce vasodilation of human omental arteries at 100 µM. Esomeprazole is safe to use in pregnancy and a candidate treatment for preeclampsia. Using primary human tissues and cells, we validated that esomeprazole is effective in enhancing vascular relaxation, and can reduce key factors associated with preeclampsia, including sFLT-1 and endothelial dysfunction. However, esomeprazole MH was more efficacious than esomeprazole MTH in our in vitro studies.


Subject(s)
Pre-Eclampsia , Biomarkers/metabolism , Esomeprazole/metabolism , Esomeprazole/pharmacology , Esomeprazole/therapeutic use , Female , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Magnesium Hydroxide , Placenta/metabolism , Pre-Eclampsia/metabolism , Pregnancy , Proton Pump Inhibitors/pharmacology , Proton Pump Inhibitors/therapeutic use , Vascular Endothelial Growth Factor Receptor-1/metabolism
5.
Patient Prefer Adherence ; 15: 1017-1026, 2021.
Article in English | MEDLINE | ID: mdl-34040355

ABSTRACT

PURPOSE: The importance of shared decision-making (SDM) between physicians and patients is increasingly recognized. In Japan, patients have shown more willingness to participate in treatment if medical professionals provide sufficient information; however, relationships between physicians and patients have traditionally been asymmetric, with patients accepting information from physicians without discussion. To explore the benefits of SDM in cancer treatment, including confidence in treatment decisions, satisfaction with treatment, and trust in healthcare providers, this study developed Japanese versions of the Control Preference Scale (CPS) and Information Needs Questionnaire (INQ). PATIENTS AND METHODS: Reliability and validity of the CPS and INQ were tested with 49 breast cancer patients. RESULTS: The CPS showed good test-retest reliability (kappa coefficient: 0.61, weighted kappa coefficient: 0.61, Kendall's tau coefficient: 0.61) and acceptable criterion validity. The INQ showed adequate consistency; the mean number of circular triads and coefficient of consistency were 3 (range 0-19) and 0.9 (range 0.37-1), respectively. Using the CPS and INQ to identify patients' roles in decision-making and information needs, results further suggested that breast cancer patients in Japan want to participate in SDM. Medical issues, including disease spread and cure, were found to be of high interest, while social and psychological issues, including sexual attractiveness, genetic risk, and family impact, tended to be low. CONCLUSION: The Japanese CPS and INQ can be used to assess patients' needs to improve care. Further, as patients' information needs change along the care trajectory, these tools should be used throughout treatment.

6.
Pharmacol Ther ; 217: 107659, 2021 01.
Article in English | MEDLINE | ID: mdl-32800789

ABSTRACT

Inappropriately activated T cells mediate autoimmune diseases and T cell acute lymphoblastic leukemia (T-ALL). Glucocorticoid and chemotherapeutic agents have largely extended lives of these patients. However, serious side effects and drug resistance often limit the prognosis of considerable number of the patients. The efficient treatment of autoimmune diseases or T-ALL with drug resistance remains an important unmet demand clinically. Bisbenzylisoquinoline alkaloids tetrandrine and cepharanthine have been applied for the treatment of certain types of autoimmune diseases and cancers, while studies on their action mechanisms and their further applications combined with glucocorticoids or chemotherapeutic agents remains to be expanded. This review introduced molecular mechanisms of tetrandrine and cepharanthine in T cells, including their therapeutic implications. Both tetrandrine and cepharnthine influence the growth of activated T cells via several kinds of signaling pathways, such as NF-κB, caspase cascades, cell cycle, MAPK, and PI3K/Akt/mTOR. According to recent preclinical and clinical studies, P-glycoprotein inhibitory effect of tetrandrine and cepharnthine could play a significant role on T cell-involved refractory diseases. Therefore, tetrandrine or cepharanthine combined with glucocorticoid or other anti-leukemia drugs would bring a new hope for patients with glucocorticoid-resistant autoimmune disease or refractory T-ALL accompanied with functional P-glycoprotein. In conclusion, bisbenzylisoquinoline alkaloids tetrandrine and cepharanthine can regulate several signaling pathways in abnormally activated T cells with low toxicity. Bisbenzylisoquinoline alkaloids deserve to be paid more attention as a lead compound to develop new drugs for the treatment of T cell-involved diseases in the future.


Subject(s)
Autoimmune Diseases/drug therapy , Benzylisoquinolines/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , T-Lymphocytes/drug effects , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Benzylisoquinolines/therapeutic use , Cell Cycle/drug effects , Down-Regulation , Drug Therapy, Combination , Glucocorticoids/therapeutic use , Humans , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/drug effects , NF-kappa B/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism
7.
Eur J Pharmacol ; 881: 173232, 2020 Aug 15.
Article in English | MEDLINE | ID: mdl-32525004

ABSTRACT

Glucocorticoids are used as anticancer and immunosuppressive agents, whereas glucocorticoid resistance has been observed in a significant fraction of patients due to overexpression of P-glycoprotein encoded by multi-drug resistance-1 gene. Tetrandrine is a bisbenzylisoquinoline alkaloid isolated from traditional herb Fangji. According to our previous report, tetrandrine potentiated glucocorticoid pharmacodynamics partially via inhibiting P-glycoprotein function. In the present study, we investigated whether glucocorticoid receptor translocation was influenced indirectly by tetrandrine via P-glycoprotein inhibition, using human T lymphoblastoid leukemia MOLT-4 cell line with little P-glycoprotein expression and its multidrug resistant sub-line MOLT-4/DNR exhibiting a large amount of P-glycoprotein. Molecular mechanism investigation suggested that overexpressed P-glycoprotein weakened the glucocorticoid receptor translocation in MOLT-4/DNR cells comparing with the parent MOLT-4 cells. Our data also suggested that tetrandrine enhanced nuclear glucocorticoid receptor translocation in MOLT-4/DNR cells indirectly by dual influences on P-glycoprotein, inhibiting the efflux function and downregulating the protein expression. Therefore, tetrandrine potentiated the cytotoxic effect of methylprednisolone against MOLT-4/DNR cells with less effects on MOLT-4 cells. These effects of tetrandrine were suggested to be beneficial for the treatment of glucocorticoid resistant diseases induced by the overexpression of P-glycoprotein.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Benzylisoquinolines/pharmacology , Daunorubicin/pharmacology , Drug Resistance, Neoplasm , Leukemia, T-Cell/drug therapy , Receptors, Glucocorticoid/metabolism , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Active Transport, Cell Nucleus , Cell Line, Tumor , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic , Humans , Leukemia, T-Cell/genetics , Leukemia, T-Cell/metabolism , Leukemia, T-Cell/pathology , Signal Transduction
8.
Oncol Rep ; 44(3): 1293, 2020 09.
Article in English | MEDLINE | ID: mdl-32582993

ABSTRACT

Following the publication of the above paper, an interested reader drew to our attention the fact that the same ß­actin bands had been included in the western blots featured in Figs. 7 and 10. Upon consulting the authors in relation to this matter, they were able to offer a legitimate explanation for this apparent duplication of the bands; essentially, in the western blotting experiments, different antibodies were used in one membrane to present different target proteins by stripping and reprobing the membrane. That the authors had performed this additional step in their protocol was inadvertently omitted from the 'Western blot analyses' subsection of the Materials and methods section. The authors are grateful to the interested reader for drawing this matter to their attention, and apologize for any inconvenience caused to the readership of the Journal. [the original article was published in Oncology Reports 41: 27-42, 2019; DOI: 10.3892/or.2018.6780].

9.
Am J Chin Med ; 47(5): 1149-1170, 2019.
Article in English | MEDLINE | ID: mdl-31311297

ABSTRACT

Three-dimensionally (3D) cultured tumor cells (spheroids) exhibit more resistance to therapeutic agents than the cells cultured in traditional two-dimensional (2D) system (monolayers). We previously demonstrated that arsenic disulfide (As2S2) exerted significant anticancer efficacies in both 2D- and 3D-cultured MCF-7 cells, whereas 3D spheroids were shown to be resistant to the As2S2 treatment. L-buthionine-(S, R)-sulfoximine (BSO), an inhibitor of glutathione (GSH) synthesis, has been regarded to be a potent candidate for combinatorial treatment due to its GSH modulation function. In the present study, we introduced BSO in combination with As2S2 at a low concentration to investigate the possible enhancing anticancer efficacy by the combinatorial treatment on 2D- and 3D-cultured MCF-7 cells. Our results presented for the first time that the combination of As2S2 and BSO exerted potent anticancer synergism in both MCF-7 monolayers and spheroids. The IC50 values of As2S2 in combinatorial treatment were significantly lower than those in treatment of As2S2 alone in both 2D- and 3D-cultured MCF-7 cells (P<0.01, respectively). In addition, augmented induction of apoptosis and enhanced cell cycle arrest along with the regulation of apoptosis- and cell cycle-related proteins, as well as synergistic inhibitions of PI3K/Akt signals, were also observed following co-treatment of As2S2 and BSO. Notably, the combinatorial treatment significantly decreased the cellular GSH levels in both 2D- and 3D-cultured MCF-7 cells in comparison with each agent alone (P<0.05 in each). Our results suggest that the combinatorial treatment with As2S2 and BSO could be a promising novel strategy to reverse arsenic resistance in human breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenicals/pharmacology , Breast Neoplasms/physiopathology , Buthionine Sulfoximine/pharmacology , Sulfides/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Culture Techniques , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Drug Synergism , Female , Humans , MCF-7 Cells , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism
10.
Anticancer Res ; 39(7): 3543-3551, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31262878

ABSTRACT

BACKGROUND/AIM: Both bevacizumab (BEV) and soluble fms-like tyrosine kinase-1 (sFlt-1) have demonstrated anti-angiogenic effects, thereby causing hypertension and proteinuria. We hypothesized that anti-preeclamptic drugs that combat the action of sFlt-1 may reduce BEV's anti-tumor efficacy. MATERIALS AND METHODS: 3D co-cultured human mini-tumors consisting of endothelial cells, fibroblasts, and cancer cells were developed. The influence of anti-preeclamptic drugs and BEV on the invasion of mini-tumors embedded in collagen gel was evaluated. RESULTS: Mini-tumor spheroids that contained MDA-MB-231 cells showed higher invasion ability than spheroids with A549. Among the six anti-preeclamptic drugs investigated, only nicorandil enhanced the invasion of mini-tumors and inhibited the action of BEV. Glibenclamide, an ATP-sensitive potassium channel inhibitor, completely quenched the action of nicorandil on mini-tumors. CONCLUSION: In the human mini-tumor model, nicorandil aggravated the invasion of mini-tumors. These data raise the possibility that concomitant use of nicorandil counteracts the efficacy of BEV therapy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antihypertensive Agents/pharmacology , Bevacizumab/pharmacology , Spheroids, Cellular/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Coculture Techniques , Female , Fibroblasts/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Nicorandil/pharmacology , Pre-Eclampsia , Pregnancy , Spheroids, Cellular/physiology , Vascular Endothelial Growth Factor A/pharmacology
11.
Chem Biol Interact ; 310: 108726, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31255635

ABSTRACT

Tetrandrine (TET) and cepharanthine (CEP) are two bisbenzylisoquinoline alkaloids isolated from the traditional herbs. Recent molecular investigations firmly supported that TET or CEP would be a potential candidate for cancer chemotherapy. Prognosis of patients with glucocorticoid resistant T cell acute lymphoblastic leukemia (T-ALL) remains poor; here we examined the anti-T-ALL effects of TET and CEP and the underlying mechanism by using the glucocorticoid resistant human leukemia Jurkat T cell line in vitro. TET and CEP significantly inhibited cell viabilities and induced apoptosis in dose- and time-dependent manner. Further investigations showed that TET or CEP not only upregulated the expression of initiator caspases such as caspase-8 and 9, but also increased the expression of effector caspases such as caspase-3 and 6. As the important markers of apoptosis, p53 and Bax were both upregulated by the treatment of TET and CEP. However, TET and CEP paradoxically increased the expression of anti-apoptotic proteins such as Bcl-2 and Mcl-1, and activated the survival protein NF-κB, leading to high expression of p-NF-κB. Cell cycle arrest at S phase accompanied by increase in the amounts of cyclin A2 and cyclin B1, and decrease in cylcin D1 amount in cells treated with TET or CEP will be another possible mechanism. During the process of apoptosis in Jurkat T cells, treatment with TET or CEP also increased the phosphorylation of JNK and p38. The PI3K/Akt/mTOR signaling pathway modification appears to play significant role in the Jurkat T cell apoptosis induced by TET or CEP. Moreover, TET and CEP seemed to downregulate the expressions of p-PI3K and mTOR in an independent way from Akt, since these two drugs strongly stimulated the p-Akt expression. These results provide fundamental insights into the clinical application of TET or CEP for the treatment of patients with relapsed T-ALL.


Subject(s)
Apoptosis/drug effects , Benzylisoquinolines/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Benzylisoquinolines/therapeutic use , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Humans , Jurkat Cells , MAP Kinase Signaling System , Phosphatidylinositol 3-Kinases/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
12.
Oncol Rep ; 41(1): 27-42, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30320388

ABSTRACT

In the present study, the antitumor effects of arsenic disulfide (As2S2) on the proliferative, survival and migratory ability of human breast cancer MCF­7 and MDA­MB­231 cells were investigated, and its potential underlying molecular mechanisms with an emphasis on cell cycle arrest, apoptosis induction, autophagy induction and reactive oxygen species (ROS) generation were determined. The results indicated that As2S2 significantly inhibited the viability, survival and migration of breast cancer cells in a dose­dependent manner. In addition, it was identified that As2S2 induced cell cycle arrest primarily at G2/M phase in the two breast cancer cell lines by regulating the expression of associated proteins, including cyclin B1 and cell division cycle protein 2. In addition to cell cycle arrest, As2S2 also triggered the induction of apoptosis in cells by activating the expression of pro­apoptotic proteins, including caspase­7 and ­8, as well as increasing the B­cell lymphoma 2 (Bcl­2)­associated X protein/Bcl­2 ratio, while decreasing the protein expression of anti­apoptotic B­cell lymphoma extra­large. In addition, As2S2 stimulated the accumulation of microtubule­associated protein 1A/1B­light chain 3 (LC3)­II and increased the LC3­II/LC3­I ratio, indicating the occurrence of autophagy. As2S2 treatment also inhibited the protein expression of matrix metalloproteinase­9 (MMP­9), but increased the intracellular accumulation of ROS in the two breast cancer cell lines, which may assist in alleviating metastasis and attenuating the progression of breast cancer. Taken together, the results of the present study suggest that As2S2 inhibits the progression of human breast cancer cells through the regulation of cell cycle arrest, intrinsic and extrinsic apoptosis, autophagy, MMP­9 signaling and ROS generation.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Arsenicals/pharmacology , Breast Neoplasms/metabolism , Sulfides/pharmacology , Apoptosis , Autophagy , Breast Neoplasms/drug therapy , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Reactive Oxygen Species/metabolism
13.
Phytother Res ; 33(1): 187-196, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30357956

ABSTRACT

Sinomenine has been used as an antirheumatic drug in China. Glucocorticoid combined with sinomenine could be an alternative therapeutic approach. In this study, we evaluated the sinomenine potential effect on glucocorticoid pharmacodynamics in vitro using a human peripheral blood mononuclear cell (PBMC) culture system. We also disclosed the possible action mechanism of sinomenine with a focus on P-glycoprotein function and glucocorticoid receptor (GR) translocation into nucleus. The median (range) of methylprednisolone IC50 values against the PBMC proliferation was 3.18 (0.45-6.81) ng/mL, whereas the median (range) IC50 values of methylprednisolone combined with 0.03, 0.3, 3, and 30 µM sinomenine were 1.85 (0.05-5.15), 0.83 (0.10-3.90), 0.56(0.09-1.62), and 0.59(0.05-1.30) ng/mL, respectively. Sinomenine significantly decreased the IC50 values of methylprednisolone and enhanced the immunosuppressive effect of methylprednisolone (p < 0.05). Sinomenine alone regulated the GR translocation in both Jurkat T cells and normal human PBMCs, and the combination of sinomenine and methylprednisolone showed stronger GR-modulatory activity than methylprednisolone alone. Thus, the additive effect of sinomenine to promote the methylprednisolone immunosuppressive efficacy was suggested to be related to nuclear GR-translocation. However, sinomenine did not significantly inhibit the P-glycoprotein function in the activated PBMCs, suggesting that sinomenine's additive effect seemed to be unrelated with the P-glycoprotein inhibition.


Subject(s)
Antirheumatic Agents/therapeutic use , Leukocytes, Mononuclear/drug effects , Morphinans/chemistry , Plant Extracts/therapeutic use , Receptors, Glucocorticoid/metabolism , Antirheumatic Agents/pharmacology , Cell Proliferation/drug effects , Humans , Plant Extracts/pharmacology
14.
Am J Cancer Res ; 8(3): 366-386, 2018.
Article in English | MEDLINE | ID: mdl-29636995

ABSTRACT

Arsenic disulfide, a major effective component of realgar, has been investigated for its anti-cancer potential and shown to have therapeutic efficacies in hematological and some solid tumors. However, its effect against breast cancer is rarely reported. In this study, we investigated the anti-cancer effects of As2S2 in human breast cancer cell lines MCF-7 and MDA-MB-231, and further elucidated its underlying mechanisms. As2S2 significantly inhibited cell viabilities, induced apoptosis, and led to cell cycle arrest in both cell lines with a dose- and time-dependent manner. As2S2 upregulated pro-apoptotic proteins like p53 and PARP in MCF-7 cells. Besides, As2S2 downregulated anti-apoptotic proteins like Bcl-2 and Mcl-1, as well as cell cycle-related proteins cyclin A2 and cyclin D1 in both cell lines. Of note, the expression level of cyclin B1 was downregulated in MCF-7 cells, whereas, upregulated in MDA-MB-231 cells. Moreover, As2S2 significantly inhibited the pro-survival signals in PI3K/Akt pathway in both cell lines. In conclusion, As2S2 inhibited cell viabilities, induced apoptosis and cell cycle arrest in both MCF-7 and MDA-MB-231 cell lines by regulating the expression of key proteins involved in related pathways. These results provide fundamental insights into the clinical application of As2S2 for treatment of patients with breast cancer.

15.
Int J Oncol ; 52(6): 1959-1971, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29620191

ABSTRACT

In China, arsenic disulfide (As2S2) has been used for the treatment of hematological malignancies. The present study aimed to evaluate the effects of As2S2 on the human breast cancer MCF­7 cell line cultured in both two­dimensional (2D) monolayers and three­dimensional (3D) spheroids to explore its therapeutic potential in breast cancer treatment. Cellular viability and the induction of apoptosis were examined with a cell counting kit­8 (CCK­8) assay and flow cytometric analysis, respectively. Alterations in the expression levels of apoptosis­associated proteins, including Bcl­2­associated X protein (Bax), B­cell lymphoma 2 (Bcl­2), p53, and caspase­7, as well as the cell survival­associated proteins, phosphatidylinositol 3­kinase (PI3K), Akt, and mammalian target of rapamycin (mTOR), were assessed by western blotting. Although a dose­dependent reduction in cell viability, which occurred in association with the induction of apoptosis triggered by the addition of 2­24 µM As2S2, was observed in both 2D­ and 3D­culture systems, 3D spheroids were less sensitive to the cytotoxic effect of As2S2 compared with the 2D cultured cells. A significant increase in the expression levels of Bax, p53, and caspase­7 was observed in treated 2D­cultured cells, whereas a similar increase in the expression levels of Bax was only confirmed in treated 3D spheroids, although there was a trend towards the increased expression of p53 and caspase­7 in the 3D spheroids. These results suggested that these molecules are closely associated with As2S2­mediated cytotoxicity in the two culture systems, and further suggested that the difference in the sensitivity to As2S2 between 2D monolayers and 3D spheroids may be attributed to the differential alterations in the expression levels of proteins associated with cell mortality. Significant downregulation of the expression levels of Bcl­2, PI3K, Akt and mTOR was observed in the two culture systems. Taken together, the results of the present study demonstrated that As2S2 inhibits cell viability and induces apoptosis in both 2D­ and 3D­ cultured MCF­7 cells, which may be associated with activation of the pro­apoptotic pathway and the inhibition of pro­survival signaling. These results have provided novel insights into clinical applications of As2S2 in the treatment of patients with breast cancer.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Arsenicals/pharmacology , Breast Neoplasms/metabolism , Cell Culture Techniques/methods , Spheroids, Cellular/drug effects , Sulfides/pharmacology , Breast Neoplasms/drug therapy , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Signal Transduction/drug effects , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism
16.
Anticancer Res ; 38(4): 2101-2108, 2018 04.
Article in English | MEDLINE | ID: mdl-29599328

ABSTRACT

BACKGROUND/AIM: Chemo-sensitivity of two-dimensional (2D) monolayers and three-dimensional (3D) spheroids of human breast cancer MCF-7 cells were investigated. MATERIALS AND METHODS: MCF-7 cells were cultured in monolayers or spheroids established using a thermo-reversible gelatin polymer, in the presence of daunorubicin, docetaxel, or As2S2 Cell proliferation was examined by a Cell Counting Kit-8 assay. RESULTS: Daunorubicin, docetaxel, and As2S2 dose-dependently decreased the MCF-7 cell proliferation in both 2D- and 3D-culture systems. The 3D spheroids were less sensitive to these agents than the 2D cultured cells. Verapamil, an inhibitor of P-glycoprotein, partially enhanced the antiproliferative effects of the agents. DL-buthionine-(S, R)-sulfoximine significantly increased (p<0.05), while N-acetyl-L-cysteine significantly inhibited the antiproliferative effects of As2S2 (p<0.003). CONCLUSION: The 3D spheroids showed less sensitivity to the antiprolliferative efficacies of anticancer agents than the 2D cultured cells. P-Glycoprotein is suggested to be partially implicated in drug resistance. Reduction of cellular glutathione level enhanced the As2S2 cytotoxicity.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Cell Culture Techniques/methods , Drug Screening Assays, Antitumor/methods , Spheroids, Cellular/pathology , Arsenicals/pharmacology , Cell Proliferation/drug effects , Daunorubicin/pharmacology , Docetaxel , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Female , Humans , MCF-7 Cells , Spheroids, Cellular/drug effects , Sulfides/pharmacology , Taxoids/pharmacology , Tissue Scaffolds/chemistry
17.
Eur J Pharmacol ; 807: 102-108, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28408143

ABSTRACT

Glucocorticoids play significant roles in treatments of inflammatory and autoimmune diseases. Some patients show a poor or absent response even to high doses of glucocorticoids. The purpose of this study was to explore whether tetrandrine combined with glucocorticoids could be a new treatment strategy to resolve glucocorticoids resistance. Information on glucocorticoids sensitivity was usually obtained through mitogen-activated human peripheral blood mononuclear cells in cell culture procedures. Thus, human peripheral blood mononuclear cells was chosen as a model to study the immunosuppressive effect of methylprednisolone combined with tetrandrine, including the possible action mechanisms. Tetrandrine decreased the IC50 value of methylprednisolone significantly, but it showed little toxic effect on the concanavalin A-activated human peripheral blood mononuclear cells. Both tetrandrine and methylprednisolone inhibited the secretion of pro-inflammatory cytokines TNFα and IL-6 significantly and the combination showed stronger inhibitory ability. Tetrandrine and/or methylprednisolone did not increase the percentage of CD4+ CD25+ Foxp3+ regulatory T cells in CD4+ T cells. However tetrandrine with or without methylprednisolone significantly inhibited the function of drug efflux pump P-glycoprotein 170 of CD4+, CD8+ T cells and lymphocytes. Tetrandrine tended to suppress the phosphorylation of mitogen-activated protein kinase and this effect was potentiated by methylprednisolone. These tetrandrine effects were suggested to be beneficial for improving the immunosuppressive efficacy of glucocorticoids. Glucocorticoids combined with tetrandrine could be a new therapeutic approach to resolve glucocorticoids-resistance possibly via inhibiting the function of P-glycoprotein and blocking mitogen-activated protein kinase signaling pathway from but not affecting on CD4+ CD25+ Foxp3+ regulatory cells.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Benzylisoquinolines/pharmacology , Glucocorticoids/pharmacology , Leukocytes, Mononuclear/drug effects , Mitogen-Activated Protein Kinases/metabolism , Mitogens/pharmacology , Adult , Cell Proliferation/drug effects , Cytokines/biosynthesis , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , Isoenzymes/metabolism , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , MAP Kinase Signaling System/drug effects , Male , Methylprednisolone/pharmacology , Phosphorylation/drug effects , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism
18.
Placenta ; 51: 1-9, 2017 03.
Article in English | MEDLINE | ID: mdl-28292463

ABSTRACT

INTRODUCTION: Preeclampsia (PE) and fetal growth restriction (FGR) are among the leading causes of perinatal morbidity and mortality. Placental insufficiency is central to these conditions. The mechanisms underlying placental insufficiency are poorly understood. Apoptosis has long been considered the only form of regulated cell death, recent research has identified an alternate process of programmed cell death known as necroptosis [1]. Necroptosis is distinct from apoptosis, relying on the deacetylase sirtuin-2 [2], receptor interacting kinases RIPK1 and 3, and the pseudokinase MLKL [3]. We aimed to determine whether these key necroptosis effector molecules were present in human placenta and whether they are differentially expressed in severe preterm (PT) PE and FGR. METHODS: PT placentas from severe early onset (<34 weeks) PE (n = 30), FGR (n = 12) and control (18) pregnancies were collected. SIRT2 and RIPK1 localization and quantitation was determined by immunohistochemistry and western blot. Immunocytochemistry was used to detect SIRT2 and RIPK1 in trophoblastic debris from first trimester, term control and PE pregnancies. Expression of SIRT2, RIPK1, RIPK3 and MLKL was examined by qPCR. RESULTS: SIRT2 and RIPK1 were localized to the syncytiotrophoblast, villous leukocytes and vasculature in all PT placentas. A significant reduction in SIRT2 protein expression in both PE and FGR placentas was identified. RIPK1 mRNA expression was significantly increased in PE placentas. Immunofluorescence identified both SIRT2 and RIPK1 in the cytotrophoblast cytoplasm. DISCUSSION: We have identified the presence of activators of necroptosis in human placenta. Interestingly, there is differential expression in major pregnancy complications. We conclude necroptosis may contribute to placental pathophysiology that underlies serious pregnancy complications.


Subject(s)
Cell Death/physiology , Fetal Growth Retardation/metabolism , Placenta/metabolism , Pre-Eclampsia/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Sirtuin 2/metabolism , Adult , Female , Fetal Growth Retardation/pathology , Humans , Placenta/pathology , Pre-Eclampsia/pathology , Pregnancy , Pregnancy Trimester, Third , Protein Kinases/genetics , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Sirtuin 2/genetics , Trophoblasts/metabolism , Young Adult
19.
Hypertension ; 69(3): 457-468, 2017 03.
Article in English | MEDLINE | ID: mdl-28115513

ABSTRACT

Preeclampsia is a severe complication of pregnancy. Antiangiogenic factors soluble fms-like tyrosine kinase-1 (sFlt-1) and soluble endoglin are secreted in excess from the placenta, causing hypertension, endothelial dysfunction, and multiorgan injury. Oxidative stress and vascular inflammation exacerbate the endothelial injury. A drug that can block these pathophysiological steps would be an attractive treatment option. Proton pump inhibitors (PPIs) are safe in pregnancy where they are prescribed for gastric reflux. We performed functional studies on primary human tissues and animal models to examine the effects of PPIs on sFlt-1 and soluble endoglin secretion, vessel dilatation, blood pressure, and endothelial dysfunction. PPIs decreased sFlt-1 and soluble endoglin secretion from trophoblast, placental explants from preeclamptic pregnancies, and endothelial cells. They also mitigated tumor necrosis factor-α-induced endothelial dysfunction: PPIs blocked endothelial vascular cell adhesion molecule-1 expression, leukocyte adhesion to endothelium, and disruption of endothelial tube formation. PPIs decreased endothelin-1 secretion and enhanced endothelial cell migration. Interestingly, the PPI esomeprazole vasodilated maternal blood vessels from normal pregnancies and cases of preterm preeclampsia, but its vasodilatory effects were lost when the vessels were denuded of their endothelium. Esomeprazole decreased blood pressure in a transgenic mouse model where human sFlt-1 was overexpressed in placenta. PPIs upregulated endogenous antioxidant defenses and decreased cytokine secretion from placental tissue and endothelial cells. We have found that PPIs decrease sFlt-1 and soluble endoglin secretion and endothelial dysfunction, dilate blood vessels, decrease blood pressure, and have antioxidant and anti-inflammatory properties. They have therapeutic potential for preeclampsia and other diseases where endothelial dysfunction is involved.


Subject(s)
Endoglin/metabolism , Endothelium, Vascular/physiopathology , Hypertension/prevention & control , Pre-Eclampsia/drug therapy , Pregnancy, Animal , Proton Pump Inhibitors/pharmacology , Vascular Endothelial Growth Factor Receptor-1/genetics , Animals , Disease Models, Animal , Endothelium, Vascular/drug effects , Female , Gene Expression Regulation/drug effects , Humans , Hypertension/metabolism , Hypertension/physiopathology , Mice , Oxidative Stress , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Pregnancy , RNA, Messenger/genetics , Trophoblasts/metabolism , Trophoblasts/pathology , Vascular Endothelial Growth Factor Receptor-1/biosynthesis , Vasodilation
20.
Immunopharmacol Immunotoxicol ; 38(5): 372-8, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27443993

ABSTRACT

CONTEXT: Diabetes mellitus is one of the most common causes of chronic renal failure. Immunosuppressive efficacies of glucocorticoids, calcineurin inhibitors, and mycophenolic acid are possibly affected by insulin after renal transplantation in these patients. OBJECTIVES: We investigated the effects of insulin on responses of mitogen-activated human peripheral blood mononuclear cells (PBMCs) to several immunosuppressive drugs. MATERIALS AND METHODS: Antiproliferative efficacies of prednisolone, hydrocortisone, cyclosporine, tacrolimus, and mycophenolic acid against concanavalin A-stimulated PBMCs were evaluated in the presence of physiological (5 µunits/mL) and super physiological (50 µunits/mL) concentrations of insulin. Insulin-receptor expressions on PBMCs were evaluated by flow cytometry. RESULTS: Insulin itself had no effects on the mitogen-induced proliferation of PBMCs. The IC50 values of cyclosporine against the mitogen-activated PBMCs in the presence of 5 or 50 µunits/mL insulin were significantly higher than those of cyclosporine without insulin (p < 0.05). The IC50 values of mycophenolic acid significantly increased by 50 µunits/mL insulin (p < 0.01). Insulin receptors were detected on the mitogen-activated CD4(+)/CD14(+ )cells in PBMCs. DISCUSSION AND CONCLUSIONS: These results indicate that insulin at even physiological concentration attenuates suppressive efficacies of several immunosuppressive drugs against mitogen-activated proliferation of human PBMCs, possibly via insulin receptors. Insulin used in dialysis patients accompanying diabetes mellitus is suggested to attenuate efficacies of immunosuppressive drugs after renal transplantation.


Subject(s)
Cell Proliferation/drug effects , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/pharmacokinetics , Insulin/pharmacology , Leukocytes, Mononuclear/immunology , Female , Humans , Leukocytes, Mononuclear/pathology , Male , Mitogens/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...