Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Mol Carcinog ; 63(8): 1611-1620, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38780147

ABSTRACT

Sulforaphane (SFN) exerts anticancer effect on various cancers including gastric cancer. However, the regulatory effect of SFN on programmed death-ligand 1 (PD-L1) and checkpoint blockade therapy in gastric cancer have not been elucidated. Here we demonstrated that SFN suppressed gastric cancer cell growth both in vitro and in vivo study. SFN upregulated PD-L1 expression through activating ΔNP63α in gastric cancer cells. Further, we found that SFN impaired the anticancer effect of anti-PD-L1 monoclonal antibody (α-PD-L1 mab) on gastric cancer cells. These results uncover a novel PD-L1 regulatory mechanism and the double-edged role of SFN in gastric cancer intervention.


Subject(s)
B7-H1 Antigen , Immune Checkpoint Inhibitors , Isothiocyanates , Stomach Neoplasms , Sulfoxides , Transcription Factors , Isothiocyanates/pharmacology , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , B7-H1 Antigen/metabolism , Sulfoxides/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Animals , Mice , Cell Line, Tumor , Transcription Factors/metabolism , Transcription Factors/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Xenograft Model Antitumor Assays , Mice, Nude
2.
Lipids Health Dis ; 23(1): 232, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080674

ABSTRACT

OBJECTIVE: To investigate how antiretroviral therapy (ART) regimens and body mass index (BMI) interact to affect triglyceride (TG) levels in people living with HIV (PLWH). METHODS: This research involved 451 men living with HIV for cross-sectional analysis, and 132 underwent follow-up assessments in 2021 and 2023. Multivariate logistic regression identified key factors, while covariance regression models assessed interactions between ART regimens and BMI on TG levels. RESULTS: The result of this cross-sectional study indicated that advanced AIDS (acquired immune deficiency syndrome) stage (OR = 2.756, P = 0.003), higher BMI (OR = 1.131, P = 0.003), and waist-hip ratio (WHR, OR = 44.684, P = 0.019) are closely associated with high triglyceride levels. Additionally, regimens containing zidovudine (AZT) (OR = 3.927, P < 0.001) or protease inhibitors/integrase strand transfer inhibitors (PI/INSTI) (OR = 5.167, P < 0.001) were significantly linked to hypertriglyceridemia. Cross-sectional and longitudinal analyses from 2021 to 2023 emphasized that changes in BMI interact with antiretroviral treatment regimens to affect TG levels in PLWH (Pinteraction < 0.05). Especially in the AZT-based drug regimen, the correlation between BMI and TG is more prominent. CONCLUSION: The interaction between ART regimens and BMI influences TG levels in PLWH, indicating that weight management is crucial for reducing the risk of hypertriglyceridemia in this population.


Subject(s)
Body Mass Index , HIV Infections , Triglycerides , Zidovudine , Humans , Male , Triglycerides/blood , Cross-Sectional Studies , HIV Infections/drug therapy , HIV Infections/blood , Longitudinal Studies , Adult , Middle Aged , Zidovudine/therapeutic use , Anti-HIV Agents/therapeutic use , Waist-Hip Ratio , Hypertriglyceridemia/blood , Antiretroviral Therapy, Highly Active
3.
Phytother Res ; 38(2): 776-796, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38050789

ABSTRACT

Clinical treatment and preclinical studies have highlighted the role of immune checkpoint blockade in cancer treatment. Research has been devoted to developing immune checkpoint inhibitors in combination with other drugs to achieve better efficacy or reduce adverse effects. Phytochemicals sourced from vegetables and fruits have demonstrated antiproliferative, proapoptotic, anti-migratory, and antiangiogenic effects against several cancers. Phytochemicals also modulate the tumor microenvironment such as T cells, regulatory T cells, and cytokines. Recently, several phytochemicals have been reported to modulate immune checkpoint proteins in in vivo or in vitro models. Phytochemicals decreased programmed cell death ligand-1 expression and synergized programmed cell death receptor 1 (PD-1) monoclonal antibody to suppress tumor growth. Combined administration of phytochemicals and PD-1 monoclonal antibody enhanced the tumor growth inhibition as well as CD4+ /CD8+ T-cell infiltration. In this review, we discuss immune checkpoint molecules as potential therapeutic targets of cancers. We further assess the impact of phytochemicals including carotenoids, polyphenols, saponins, and organosulfur compounds on cancer PD-1/programmed cell death ligand-1 immune checkpoint molecules and document their combination effects with immune checkpoint inhibitors on various malignancies.


Subject(s)
B7-H1 Antigen , Neoplasms , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Programmed Cell Death 1 Receptor/metabolism , Immune Checkpoint Proteins , Ligands , Immunotherapy , Neoplasms/drug therapy , Antibodies, Monoclonal/therapeutic use , Tumor Microenvironment
4.
Mol Carcinog ; 62(11): 1673-1685, 2023 11.
Article in English | MEDLINE | ID: mdl-37477518

ABSTRACT

Gastric cancer is one of the deadliest malignant tumors, and half of the patients develop recurrences or metastasis within 5 years after eradication therapy. Cancer stem cells (CSCs) are considered to be important in this progress. The sonic hedgehog (SHH) pathway plays an important role in the maintenance of gastric CSCs characteristics. The p63 proteins are vital transcription factors belonging to the p53 family, while their functions in regulating CSCs remain unclear. The preventive effects of dietary diallyl trisulfide (DATS) against human gastric cancer have been verified. However, whether DATS can target gastric CSCs are poorly understood. Here, we investigated the role of ΔNp63/SHH pathway in gastric CSCs and the inhibitory effect of DATS on gastric CSCs via ΔNp63/SHH pathway. We found that ΔNp63 was upregulated in serum-free medium cultured gastric tumorspheres compared with the parental cells. Overexpression of ΔNp63 elevated the self-renewal capacity and CSC markers' levels in gastric sphere-forming cells. Furthermore, we found that ΔNp63 directly bound to the promoter region of Gli1, the key transcriptional factor of SHH pathway, to enhance its expression and to activate SHH pathway. In addition, it was revealed that DATS effectively inhibited gastric CSC properties both in vitro and in vivo settings. Activation of SHH pathway attenuated the suppressive effects of DATS on the stemness of gastric cancer. Moreover, DATS suppression of gastric CSC properties was also diminished by ΔNp63 upregulation through SHH pathway activation. These findings illustrated the role of ΔNp63/SHH pathway in DATS inhibition of gastric cancer stemness. Taken together, the present study suggested for the first time that DATS inhibited gastric CSCs properties by ΔNp63/SHH pathway.


Subject(s)
Hedgehog Proteins , Stomach Neoplasms , Humans , Hedgehog Proteins/metabolism , Hedgehog Proteins/pharmacology , Stomach Neoplasms/pathology , Signal Transduction , Transcription Factors/metabolism , Neoplastic Stem Cells/pathology , Cell Line, Tumor
5.
Nutr Cancer ; 75(3): 980-991, 2023.
Article in English | MEDLINE | ID: mdl-36542459

ABSTRACT

Sulforaphane (SFN), a major isothiocyanate found in cruciferous vegetables, reportedly exerts extensive antitumor effects. Butyl benzyl phthalate (BBP), a widely used plasticizer, plays a crucial role in the promotion of breast cancer. In the present study, we demonstrated that SFN inhibited proliferation, induced apoptosis, and suppressed the stemness of MCF-7 cells, whereas BBP exerted the opposite effects; microRNA-19 (miR-19) plays an important role in BBP-induced cell growth and dysregulation mediated via PTEN and p21. The growth-promoting effect of BBP could be mitigated by SFN, accompanied by a reversal of altered expression of miR-19a, miR-19b, PTEN, and p21. SFN also suppressed BBP-induced binding of upregulated miR-19 with PTEN, as determined using a dual-luciferase reporter assay. Collectively, these results demonstrated, for the first time, that SFN regulates the miR-19/PTEN axis to exert protective effects against BBP-mediated breast cancer promotion, suggesting a new potential role for SFN (or SFN-rich foods) in phthalate antagonism.


Subject(s)
Breast Neoplasms , MicroRNAs , Humans , Female , MicroRNAs/genetics , MicroRNAs/metabolism , MCF-7 Cells , Breast Neoplasms/pathology , Isothiocyanates/pharmacology , Cell Proliferation , Apoptosis , Cell Line, Tumor , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism
6.
Nutr Cancer ; 75(3): 971-979, 2023.
Article in English | MEDLINE | ID: mdl-36562732

ABSTRACT

Cancer stem-like cells (CSCs), which play an important role in tumor initiation and progression, have been identified in many cancers. Diallyl trisulfide (DATS) is an organosulfur compound extracted from garlic with anticancer activities. Nanog is a transcription factor responsible for maintaining the stemness of CSCs, but its role in the DATS-induced attenuation of renal CSC properties is unknown. In this study, renal CSCs were enriched from human renal cancer cell lines 786-O and ACHN cultured in a serum-free medium (SFM). The properties of CSCs were analyzed by evaluating the ability of the cells in sphere formation and measuring the expression of stem cell markers. We found that downregulation of Nanog inhibited renal CSC properties. DATS suppressed renal CSC activities by reducing tumorsphere formation, decreasing stem cell markers including Nanog, CD44, ALDH1A1, and Oct4, inhibiting cell proliferation and promoting apoptosis. We further revealed that overexpression of Nanog reversed the suppressive effects of DATS on renal CSCs. Taken together, our results demonstrated that DATS inhibited renal CSCs by suppressing Nanog. These novel findings suggested that, through Nanog targeting, DATS can potentially be used as an anti-tumor agent for renal cancer.


Subject(s)
Allyl Compounds , Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Cell Line, Tumor , Sulfides/pharmacology , Allyl Compounds/pharmacology , Carcinoma, Renal Cell/pathology , Apoptosis , Kidney Neoplasms/pathology , Neoplastic Stem Cells , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Nanog Homeobox Protein/pharmacology
7.
Ecotoxicol Environ Saf ; 252: 114605, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36753971

ABSTRACT

BACKGROUND: The omnipresence of human phthalate (PAE) exposure is linked to various adverse health issues, including breast cancer. However, the effects of low-dose PAE exposure on breast cancer stem cells (BCSCs) and the underlying mechanism remain unexplored. METHODS: BCSCs from breast cancer cell lines (MDA-MB-231 and MCF-7) were enriched using a tumorsphere formation assay. Gene and protein expression was detected by measurement of quantitative real-time reverse transcription PCR, western blot, and immunofluorescence assays. Transient transfection assays were used to evaluate the involvement of Gli1, a signaling pathway molecule and ΔNp63α, an oncogene in influencing the PAE-induced characteristics of BCSCs. RESULTS: PAE (butylbenzyl phthalate, BBP; di-butyl phthalate, DBP; di-2-ethylhexyl phthalate, DEHP) exposure of 10-9 M significantly promoted the tumorsphere formation ability in BCSCs. Breast cancer spheroids with a 10-9 M PAE exposure had higher levels of BCSC marker mRNA and protein expression, activated sonic hedgehog (SHH) pathway, and increased mRNA and protein levels of an oncogene, ΔNp63α. Furthermore, suppression of the SHH pathway attenuated the effects of PAEs on BCSCs. And the overexpression of ΔNp63α enhanced PAE-induced characteristics of BCSCs, while low expression of ΔNp63α inhibited the promotion effects of PAEs on BCSCs and the SHH pathway. CONCLUSION: Low-dose PAE exposure promoted the stem cell properties of BCSCs in a ΔNp63α- and SHH-dependent manner. The influence of low-dose exposure of PAEs and its relevance for the lowest observed effect concentrations requires further investigation, and the precise underlying mechanism needs to be further explored.


Subject(s)
Breast Neoplasms , Hedgehog Proteins , Humans , Female , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Signal Transduction , Oncogenes , Neoplastic Stem Cells/metabolism , Cell Line, Tumor
8.
Phytother Res ; 37(7): 2995-3008, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36866538

ABSTRACT

Resveratrol (RES) has various pharmacological bioactivities and its anticancer effects in lung cancer have been proven. However, the underlying mechanisms of action of RES in lung cancer remain unclear. This study focused on Nrf2-mediated antioxidant systems in RES-treated lung cancer cells. A549 and H1299 cells were treated with various concentrations of RES at different times. RES decreased cell viability, inhibited cell proliferation, and increased the number of senescent and apoptotic cells in a concentration- and time-dependent manner. Moreover, RES-induced lung cancer cell arrest at the G1 phase was accompanied by changes in apoptotic proteins (Bax, Bcl-2, and cleaved caspase 3). Furthermore, RES induced a senescent phenotype along with changes in senescence-related markers (senescence-associated ß-galactosidase activity, p21, and p-γH2AX). More importantly, with prolonged exposure time and increased exposure concentration, intracellular reactive oxygen species (ROS) continuously accumulated, resulting in a decrease in Nrf2 and its downstream antioxidant response elements, including CAT, HO-1, NQO1, and SOD1. Meanwhile, RES-induced ROS accumulation and cell apoptosis were reversed by N-acetyl-l-cysteine treatment. Taken together, these results suggest that RES disturb lung cancer cellular homeostasis by destroying the intracellular antioxidant pool to increase ROS production. Our findings provide a new perspective on RES intervention in lung cancer.


Subject(s)
Antioxidants , Lung Neoplasms , Humans , Antioxidants/pharmacology , Resveratrol/pharmacology , Reactive Oxygen Species/metabolism , NF-E2-Related Factor 2/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Apoptosis , Cellular Senescence , Cell Line, Tumor
9.
Cell Biol Toxicol ; 38(2): 273-289, 2022 04.
Article in English | MEDLINE | ID: mdl-33811578

ABSTRACT

Interleukin-17A (IL-17A) is an essential inflammatory cytokine in the progress of carcinogenesis. Tobacco smoke (TS) is a major risk factor of lung cancer that influences epithelial-mesenchymal transition (EMT) process. However, the potential mechanism by which IL-17A mediates the progression of lung cancer in TS-induced EMT remains elusive. In the present study, it was revealed that the IL-17A level was elevated in lung cancer tissues, especially in tumor tissues of cases with experience of smoking, and a higher IL-17A level was correlated with induction of EMT in those specimens. Moreover, the expression of ΔNp63α was increased in IL-17A-stimulated lung cancer cells. ΔNp63α functioned as a key oncogene that bound to the miR-17-92 cluster promoter and transcriptionally increased the expression of miR-19 in lung cancer cells. Overexpression of miR-19 promoted EMT in lung cancer with downregulation of E-cadherin and upregulation of N-cadherin, while its inhibition suppressed EMT. Finally, the upregulated levels of IL-17A, ΔNp63α, and miR-19 along with the alteration of EMT-associated biomarkers were found in lung tissues of TS-exposed mice. Taken together, the abovementioned results suggest that IL-17A increases ΔNp63α expression, transcriptionally elevates miR-19 expression, and promotes TS-induced EMT in lung cancer. These findings may provide a new insight for the identification of therapeutic targets for lung cancer.


Subject(s)
Lung Neoplasms , MicroRNAs , Tobacco Smoke Pollution , Animals , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Interleukin-17/genetics , Interleukin-17/metabolism , Lung Neoplasms/pathology , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Smoke , Nicotiana/metabolism
10.
Bioorg Chem ; 128: 106034, 2022 11.
Article in English | MEDLINE | ID: mdl-35908353

ABSTRACT

Liver disease has become a major cause of premature mortality worldwide. It is well known that dysregulated inflammation response plays a crucial role in most liver diseases. As a Chinese medicinal herb, Magnesium isoglycyrrhizinate (MgIG) has been proven to have good hepatoprotective activity and has been used in clinic to treat liver disease. However, the mechanisms by which MgIG regulates LPS-induced liver injury and inflammation in vivo remain elusive. In our study, MgIG pretreatment mitigated LPS-induced liver damage by suppressing apoptosis and inflammation via regulating macrophage/neutrophil infiltration. MgIG ameliorated the effects of LPS on pro-oxidant enzymes (NOX1/2/4) and anti-oxidant enzymes (SOD1/2). Interestingly, we found that the level of the hepatoprotective cytokine interleukin (IL)-22 was significantly upregulated in MgIG-treated liver tissues, which might be a potential mechanism of MgIG against liver injury. Moreover, we found that MgIG treatment not only inhibited TLR4/MyD88/NF-κB signaling pathway, but also activated autophagy. Furthermore, IL-22 treatment activated autophagy and inhibited TLR4/NF-κB signaling pathway in vitro, suggesting that IL-22-activated autophagy and -inhibited inflammation also participated in the protective effects of MgIG. Altogether, our results uncovered the potential mechanisms of the hepatoprotective effects of MgIG, which provided critical evidence to support the use of MgIG to prevent and treat liver diseases.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , Saponins , Triterpenes , Animals , Autophagy , Chemical and Drug Induced Liver Injury, Chronic/metabolism , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Interleukins/metabolism , Interleukins/pharmacology , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Liver , Mice , NF-kappa B/metabolism , Saponins/metabolism , Saponins/pharmacology , Saponins/therapeutic use , Toll-Like Receptor 4/metabolism , Triterpenes/metabolism , Triterpenes/pharmacology , Interleukin-22
11.
Nutr Cancer ; 73(7): 1157-1167, 2021.
Article in English | MEDLINE | ID: mdl-32586140

ABSTRACT

AIMS: Renal cell cancers typically exhibit high metastasis and recurrence, and this is thought to be due to renal cancer stem cells (CSCs). Meanwhile, aberrant activation of Sonic hedgehog (Shh) signaling is linked with CSCs. Resveratrol has direct or indirect impacts on the pathological activities of CSCs. However, the effects of resveratrol on renal CSCs remain to be elucidated. METHODS: We cultured renal CSCs in serum-free medium. Western blotting was used to analyze the expression levels of related proteins. The mRNA changes were detected by qRT-PCR after resveratrol treatment. The CD133+ cells were quantified by flow cytometry analysis. Immunofluorescence staining images showed the changes in CD44 and Smoothened expression in the cell spheres. RESULTS: Renal CSCs were enriched by tumorsphere formation assays of ACHN and 786-O cells. Resveratrol treatments markedly decreased the size and number of cell spheres and downregulated the expression of the Shh pathway-related proteins and CSCs markers. Moreover, we observed that resveratrol inhibited cell proliferation and induced cell apoptosis, while purmorphamine upregulated the Shh pathway and weakened the effects of resveratrol on renal CSCs. CONCLUSIONS: These results suggested that resveratrol is a potential novel therapeutic agent that targets inactivation of renal CSCs by affecting the function of the Shh pathway.


Subject(s)
Hedgehog Proteins , Neoplasms , Neoplastic Stem Cells/drug effects , Resveratrol , Signal Transduction , Cell Proliferation , Cells, Cultured , Humans , Resveratrol/pharmacology
12.
Environ Toxicol ; 35(11): 1274-1283, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32649042

ABSTRACT

Nanog plays an important role in the regulation of cancer stem cells (CSCs) which participate in tumorgenesis and progression. In renal cancer, tobacco smoke (TS) is considered a major risk factor. However, the molecular mechanism by which TS induces the development of renal CSC properties remains largely unknown. In this study, we showed that the level of Nanog was elevated in renal cell carcinoma (RCC) patients with a smoking history, and that Nanog overexpression promoted the traits of CSCs in renal cancer. We further demonstrated that a 8-week exposure of TS enhanced the formation of renal tumorspheres, increased the population of CD133-positive cells, and stimulated the expression of Nanog and CSC markers. In addition, TS was found to play a role in accelerating the cell growth transition from G1 to S phase in renal CSCs. Finally, we demonstrated that the TS-induced effects in renal CSCs could be reversed through the downregulation of Nanog. Our results suggested that Nanog plays a role in mediating TS-induced renal CSC properties. This study may provide new insights into the molecular mechanism of TS-related renal tumorigenesis, which can contribute to the future development of therapeutics for renal cancer.


Subject(s)
Carcinoma, Renal Cell/metabolism , Nanog Homeobox Protein/metabolism , Tobacco Smoke Pollution , Carcinogenesis/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/drug effects , Down-Regulation , Humans , Kidney Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Nicotiana/metabolism
13.
J Cell Biochem ; 120(9): 15616-15624, 2019 09.
Article in English | MEDLINE | ID: mdl-31042325

ABSTRACT

Curcumin is a phytochemical which exhibits significant inhibitory effect in multiple cancers including prostate cancer. MicroRNA-34a (miR-34a) was found to be a master tumor suppressor miRNA and regulated the growth of cancer cells. To date, however, the role of miR-34a in the anticancer action of curcumin against prostate cancer has been rarely reported. In the present study, we showed that curcumin altered the expression of cell cycle-related genes (cyclin D1, PCNA, and p21) and inhibited the proliferation of prostate cancer cells. Furthermore, we found that curcumin significantly upregulated the expression of miR-34a, along with the downregulated expression of ß-catenin and c-myc in three prostate cancer cell lines. Inhibition of miR-34a activated ß-catenin/c-myc axis, altered cell cycle-related genes expression and significantly suppressed the antiproliferation effect of curcumin in prostate cancer cells. Findings from this study revealed that miR-34a plays an important role in the antiproliferation effect of curcumin in prostate cancer.


Subject(s)
Cell Proliferation/drug effects , Curcumin/pharmacology , MicroRNAs/genetics , Prostatic Neoplasms/drug therapy , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Prostate/drug effects , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays
14.
Bioorg Med Chem ; 27(3): 516-524, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30617018

ABSTRACT

Magnesium Isoglycyrrhizinate (MgIG), a novel molecular compound extracted from licorice root, has exhibited greater anti-inflammatory activity and hepatic protection than glycyrrhizin and ß-glycyrrhizic acid. In this study, we investigated the anti-inflammatory effect and the potential mechanism of MgIG on Lipopolysaccharide (LPS)-treated RAW264.7 cells. MgIG down-regulated LPS-induced pro-inflammatory mediators and enzymes in LPS-treated RAW264.7 cells, including TNF-α, IL-6, IL-1ß, IL-8, NO and iNOS. The generation of reactive oxygen species (ROS) in LPS-treated RAW264.7 cells was also reduced. MgIG attenuated NF-κB translocation by inhibiting IKK phosphorylation and IκB-α degradation. Simultaneously, MgIG also inhibited LPS-induced activation of MAPKs, including p38, JNK and ERK1/2. Taken together, these results suggest that MgIG suppresses inflammation by blocking NF-κB and MAPK signaling pathways, and down-regulates ROS generation and inflammatory mediators.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Mitogen-Activated Protein Kinases/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Saponins/pharmacology , Triterpenes/pharmacology , Animals , Cells, Cultured , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Dose-Response Relationship, Drug , Inflammation/drug therapy , Inflammation/metabolism , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinases/metabolism , Molecular Structure , NF-kappa B/metabolism , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , RAW 264.7 Cells , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Structure-Activity Relationship
15.
Phytother Res ; 33(10): 2783-2791, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31342620

ABSTRACT

Genistein, a soy derived isoflavanoid compound, exerts anticancer effects in various cancers. Nasopharyngeal cancer stem cells (NCSCs) are a small subpopulation of cancer cells which are responsible for initiation, progression, metastasis, and recurrence of nasopharyngeal cancer. The present study aimed to investigate the suppressive effects of genistein on NCSCs and its underlying mechanism. NCSCs were enriched from human nasopharyngeal cancer cell lines CNE2 and HONE1 through tumorsphere-forming assay. It was shown that genistein inhibited the tumorsphere formation capacity, decreased the number of EpCAM+ cells, downregulated the expression of NCSCs markers, suppressed cell proliferation, and induced apoptosis of NCSCs. Genistein suppressed the activity of Sonic hedgehog (SHH) signaling, which was important for the maintenance of NCSCs, while activation of SHH signaling by purmorphamine diminished the inhibitory effects of genistein on NCSCs. Our data suggested that genistein inhibited NCSCs through the suppression of SHH signaling. These findings support the use of genistein for targeting NCSCs.


Subject(s)
Genistein/pharmacology , Hedgehog Proteins/physiology , Nasopharyngeal Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Cell Line, Tumor , Humans , Signal Transduction/drug effects
16.
Int J Food Sci Nutr ; 70(5): 570-578, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30624124

ABSTRACT

Sulforaphane (SFN) is the major component extracted from broccoli/broccoli sprouts. It has been shown to possess anti-cancer activity. Gastric cancer is common cancer worldwide. The objective of this work was to evaluate the inhibitory effect of SFN on gastric cancer by Sonic hedgehog (Hh) Pathway. The results found that tumorsphere formation and the expression levels of gastric cancer stem cells (CSCs) markers were significantly decreased after SFN treatment. SFN also exerted inhibitory effects by suppressing proliferation and inducing apoptosis in gastric CSCs. Intriguingly, SFN inhibited the activation of Sonic Hh, a key pathway in maintaining the stemness of gastric CSCs. Upregulation of Sonic Hh pathway diminished the inhibitory effects of SFN on gastric CSCs. Collectively, these data revealed that SFN could be a potent natural compound targeting gastric CSCs via suppression of Sonic Hh pathway, which might be an promising agent for gastric cancer intervention.


Subject(s)
Antineoplastic Agents/pharmacology , Hedgehog Proteins/metabolism , Isothiocyanates/pharmacology , Neoplastic Stem Cells/drug effects , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Brassica/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors , Mice , Mice, Transgenic , Sulfoxides , Up-Regulation
17.
J Cell Biochem ; 119(5): 4134-4141, 2018 05.
Article in English | MEDLINE | ID: mdl-29243835

ABSTRACT

Cancer stem cells (CSCs) play a central role in the development of breast cancer. The canonical Wnt/ß-catenin signal pathway is critical for maintaining CSCs characteristics. Diallyl trisulfide (DATS), a natural organosulfur compound from the garlic, exhibits effective antitumor properties. However, the role of DATS in regulating breast CSCs activity and the underlying molecular mechanisms remain obscure. In the present study, we reported that DATS efficiently inhibited the viability of breast CSCs as evidenced by reducing turmorspheres formation, decreasing the expression of breast CSCs markers (CD44, ALDH1A1, Nanog, and Oct4), as well as inhibiting proliferation and inducing apoptosis. Furthermore, we showed that DATS downregulated the activity of Wnt/ß-catenin pathway, while LiCl-triggered Wnt/ß-catenin activation diminished DATS inhibition on breast CSCs. Taken together, our results illustrated that DATS suppressed breast CSCs through inhibiting Wnt/ß-catenin pathway activation. These novel findings could provide new insights into the molecular mechanisms of breast CSCs regulation as well as its target intervention and might provide new strategies for preventing and treating breast cancers.


Subject(s)
Allyl Compounds/pharmacology , Breast Neoplasms/drug therapy , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Sulfides/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Antigens, Differentiation/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Humans , MCF-7 Cells , Neoplastic Stem Cells/pathology
18.
Biochem Biophys Res Commun ; 495(3): 2139-2144, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29258820

ABSTRACT

Besides its well-established oncosuppressor activity, the role of p53 in regulating metabolic pathways has been recently identified. Nevertheless, the function of p53 with respect to insulin resistance appears highly controversial. To address this issue, we investigated the expression of p53 in experimental model of insulin resistance. Then we used activator (nutlin-3α) and inhibitor (pifithrin-α, PFT-α) of p53 in HepG2 cell. Here we showed that p53 protein level was decreased in the hepatic tissue of high-fat diet-induced insulin resistance mice, genetically diabetic ob/ob mice and palmitate (PA) treated HepG2 cells. And high expression of phosphor-p38, ERK1/2 and nuclear factor kappa B (NF-κB) p65 accompanied with low expression of p53. But activation of p53 with nutlin-3α prevented PA-induced reduction of glucose consumption and suppression of insulin signaling pathways. At the same time, nutlin-3α downregulated the activation of NF-κB, p38 and ERK1/2 pathways upon stimulation with PA. In contrast, inhibition of p53 with PFT-α decreased glucose consumption and suppressed insulin signaling pathway. Furthermore, PFT-α activated NF-κB, p38 and ERK1/2 pathways in HepG2 cells. Overall, these results suggest that p53 is involved in improving insulin sensitivity of hepatic cells via inhibition of mitogen-activated protein kinases (MAPKs) and NF-κB pathways.


Subject(s)
Insulin Resistance/physiology , Liver/metabolism , MAP Kinase Signaling System/physiology , NF-kappa B/metabolism , Tumor Suppressor Protein p53/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
19.
Nutr Cancer ; 70(2): 221-228, 2018.
Article in English | MEDLINE | ID: mdl-29313724

ABSTRACT

We retrospectively examined a large cohort of esophageal carcinoma patients who received early enteral nutrition (EEN) to clarify the validity of EEN compared with total parenteral nutrition (TPN). Included were a total of 665 consecutive patients with histologically confirmed carcinoma of the esophagus or esophagogastric junction; and all patients underwent esophagectomy. The patients were divided into two groups: TPN (n = 262) and EEN (n = 403). The TPN group consisted of patients who only received intravenous nutrition support after operation. The postoperative length of hospital stay (PLOS), anastomotic leakage, mortality after surgery, and hospital charges were reviewed and analyzed. Compared with the TPN group, the EEN group had significantly shorter mean PLOS (15.6 days vs. 22.5 days; P < 0.01). Multivariable linear regression analysis revealed EEN to be associated with shorter PLOS even after adjustment for tumor histology, tumor location, type of esophagectomy, and postoperative albumin infusion. Hospital charges were also significantly less for those in the EEN group than the TPN group. There was no significant difference between the two groups regarding the complication of anastomotic leakage and clinical outcome after surgery. These findings suggest that EEN reduces PLOS and hospital charges of Chinese esophageal cancer patients who had an esophagectomy.


Subject(s)
Enteral Nutrition , Esophageal Neoplasms/surgery , Esophagectomy , Adult , Aged , Aged, 80 and over , Albumins/administration & dosage , Asian People , Cohort Studies , Esophageal Neoplasms/therapy , Female , Hospitalization/economics , Humans , Male , Middle Aged , Parenteral Nutrition/methods , Postoperative Care , Retrospective Studies , Treatment Outcome
20.
Anticancer Drugs ; 29(3): 208-215, 2018 03.
Article in English | MEDLINE | ID: mdl-29356693

ABSTRACT

Cancer stem cells (CSCs) play an essential role in the progression of many tumors. Sonic hedgehog (Shh) and Wnt/ß-catenin pathways are crucial in maintaining the stemness of CSCs. Curcumin has been shown to possess anticancer activity. However, the interventional effect of curcumin on breast CSCs has not been elucidated. In the present study, we investigated the role of Shh and Wnt/ß-catenin pathway in curcumin inhibition of breast CSCs. We showed that the levels of breast CSCs markers were significantly elevated in SUM159 and MCF7 sphere-forming cells. We further illustrated that curcumin effectively decreased breast CSCs activity by inhibiting tumor sphere formation, decreasing breast CSCs markers (CD44, ALDH1A1, Nanog, and Oct4), as well as inhibiting proliferation and inducing apoptosis. Moreover, we showed that downregulation of Shh and Wnt/ß-catenin activity resulted in breast CSCs inhibition; curcumin exerted an inhibitory effect on breast CSCs by suppressing both Shh and Wnt/ß-catenin pathways. Taken together, these results indicated curcumin inhibition of breast CSCs by downregulation of Shh and Wnt/ß-catenin pathways. Findings from this study could provide new insights into the potential therapeutic application of curcumin in breast CSCs elimination and cancer intervention.


Subject(s)
Breast Neoplasms/drug therapy , Curcumin/pharmacology , Hedgehog Proteins/metabolism , Neoplastic Stem Cells/drug effects , Wnt Signaling Pathway/drug effects , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation , Female , Humans , MCF-7 Cells , Neoplastic Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL