Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
J Biochem Mol Toxicol ; 38(2): e23642, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38348710

ABSTRACT

Breast cancer is the leading cause of cancer deaths in women worldwide. EF-24, an analog of curcumin, has been shown to possess promising anticancer effects. However, the underlying mechanism remains elusive. In the present study, the inhibitory effect of EF-24 against one breast cancer cell line, MDA-MB-231, and its anti-migration ability were assessed by MTT, wound healing, and Transwell assay. Furthermore, we found that EF-24 could induce initiation of autophagy as evidenced by fluorescence and electron microscope observation. EF-24 also induced mitochondrial apoptosis in MDA-MB-231 cells as detected by Hoechst 33342 staining, flow cytometry analysis, and western blot analysis. In addition, the early autophagy inhibitor 3-MA could reduce the cleavage of PARP protein and protect cells from EF-24-induced apoptosis, while the autophagy inducer (rapamycin) could enhance the anticancer effect of EF-24 in MDA-MB-231 cells, which suggest that EF-24 induces crosstalk between autophagy and apoptosis, which herein participate in the antiproliferative effect of EF-24 in breast cancer cells. Moreover, removal of EF-24-activated ROS with NAC significantly reversed migration ability of MDA-MB-231 cells, indicating that EF-24 exerted an inhibitory effect through a ROS-mediating pathway. These results will help to elucidate the antitumor mechanism of curcumin analogs and to explore future potential clinical applications.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Curcumin , Female , Humans , Curcumin/pharmacology , Curcumin/therapeutic use , MDA-MB-231 Cells , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Reactive Oxygen Species/metabolism , Cell Proliferation , Breast Neoplasms/pathology , Autophagy , Apoptosis , Cell Line, Tumor
2.
J Enzyme Inhib Med Chem ; 39(1): 2339901, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38864175

ABSTRACT

The spices and aromatic herbs were used not only in cooking to add flavour and smell to dishes but also for medicinal use. Nigella sativa, also called black cumin, is one of the species that contains an important bioactive component, thymoquinone (TQ), which has antioxidant, anti-inflammatory, antimicrobial, and antidiabetic effects. Curcuma longa, which also includes curcumin, has numerous anti-cancer properties. However, the bioavailability of curcumin is lower than that of its analogs. An analog of curcumin (EF-24), which has better bioavailability than curcumin, is capable of exerting a high anti-cancer effect. In our study, we determined the effects of PON1 enzyme activity on the proliferation and aggressiveness of glioblastoma cancer treated with TQ and EF-24 from lysates of the glioblastoma cell line U87MG. The results were determined as increased PON1 activity after treatment with TQ and EF-24 in the U87MG cell line (p < 0.0001).


Subject(s)
Aryldialkylphosphatase , Benzoquinones , Cell Proliferation , Curcumin , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Glioblastoma , Humans , Aryldialkylphosphatase/metabolism , Aryldialkylphosphatase/antagonists & inhibitors , Glioblastoma/drug therapy , Glioblastoma/pathology , Benzoquinones/pharmacology , Benzoquinones/chemistry , Curcumin/pharmacology , Curcumin/chemistry , Curcumin/chemical synthesis , Cell Proliferation/drug effects , Molecular Structure , Structure-Activity Relationship , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Tumor Cells, Cultured
3.
Saudi Pharm J ; 32(1): 101892, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38146327

ABSTRACT

Pentagamavunone-1 (PGV-1), an analog of curcumin, has been studied for its cytotoxic effects in 4T1, MCF7, MCF7/HER2, and T47D breast cancer cells. Its antiproliferative effect is partly mediated through G2/M arrest; however, its molecular mechanism during cell cycle progression remains unknown. In this study, we aimed to determine whether PGV-1 has any anticancer effects on highly aggressive breast cancer cells, with a focus on cell cycle regulatory activity, reactive oxygen species (ROS) generation, and their mediated effects on cancer cells. MDA-MB-231 (triple-negative) and HCC1954 (overexpressed HER2) immortalized human breast cancer cells were used in the study. PGV-1 exhibited cytotoxic activity with an irreversible antiproliferative impact on treated cells and had good selectivity when tested in fibroblast cells. Oral PGV-1 administration suppressed tumor growth in a cell-derived xenograft mouse model. PGV-1 induced the phosphorylation of Aurora A kinase and PLK1 in MDA-MB-231 cells, while PLK1 and cyclin B1 phosphorylation were enhanced in the PGV-1-treated HCC1954 cells during prometaphase arrest. Intracellular ROS production was substantially higher upon PGV-1 treatment following mitotic arrest, and this activity caused impairment of mitochondrial respiration, induced senescence, and subsequently triggered early-to-late apoptosis. Collectively, these results suggest that the molecular mechanism of PGV-1 involves the regulation of mitotic kinases to cause cell cycle arrest and the enhancement of ROS production to impair mitochondrial activity and induce cellular senescence. The therapeutic activities demonstrated by PGV-1 in this study show its potential as an appealing candidate for chemotherapy in breast cancer treatment.

4.
J Biochem Mol Toxicol ; 37(4): e23307, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36633067

ABSTRACT

5-Bis[(2-fluorophenyl)methylene]-4-piperidinone (EF-24) is a curcumin analog, which was identified for its physiochemical stability and diverse pharmacological functions. In the present study, EF-24 was added to the breast cancer cell line MCF-7 and its cellular effects were characterized. The results indicated that EF-24 possessed antiproliferative and antimigratory activities on MCF-7 cells as determined by MTT assay, wound healing, and transwell assay, respectively. In addition, the autophagosomal vesicles could be detected by acridine orange staining and electron microscope analysis in EF-24-treated cells. Conversion of LC3-I to LC3-II was also investigated following EF-24 treatment of the cells. However, the expression analysis of p62 and LC3 revealed that EF-24 could inhibit autophagic flux in MCF-7 cells. Confocal microscopy suggested that EF-24 could inhibit the degradation of autophagic vesicles by blocking the fusion of autophagosomes with lysosomes. EF-24 could also induce apoptosis of MCF-7 cells as determined by Hoechst 33342 staining, flow cytometry analysis, and western blot analysis. Moreover, treatment of the cells with the autophagy inhibitor 3-MA enhanced the PARP1 cleavage of EF-24-treated MCF-7 cells, which indicated the crosstalk between autophagy and apoptosis in breast cancer cells. Additional investigation of EF-24 should be performed in future studies to assess its antiproliferation and antimigratory effects on MCF-7 cells. However, the current results provide a solid foundation for the potential in vivo anticancer activity of this compound.


Subject(s)
Breast Neoplasms , Curcumin , Humans , Female , MCF-7 Cells , Curcumin/pharmacology , Cell Proliferation , Autophagy , Cell Line, Tumor , Breast Neoplasms/drug therapy , Apoptosis
5.
Biol Pharm Bull ; 46(6): 756-763, 2023.
Article in English | MEDLINE | ID: mdl-37258140

ABSTRACT

Curcumin has long been recognized for its anti-inflammatory properties. An antitumor effect has been recently reported in curcumin and clinical trials are being conducted. However, a large amount of required intake to obtain the antitumor effect of curcumin has been regarded as a problem. Therefore, curcumin analogs have been created by many researchers to enhance the effects of curcumin. We have synthesized >50 curcumin analogs and revealed greater growth suppression of various tumor cells with mono-carbonyl analogs than curcumin. Mechanistically, mono-carbonyl analogs inhibited transcriptional activity (e.g., nuclear factor kappa B, signal transducer, and activator of transcription 3) or activated caspase-3. Additionally, mono-carbonyl analogs of curcumin control tumor cell metabolism. Herein, we summarize the current knowledge about mono-carbonyl curcumin analogs and discuss their potential clinical applications.


Subject(s)
Curcumin , Neoplasms , Humans , Curcumin/pharmacology , Curcumin/therapeutic use , Neoplasms/drug therapy , Anti-Inflammatory Agents/pharmacology , NF-kappa B/metabolism , Cell Line, Tumor
6.
J Asian Nat Prod Res ; 25(5): 456-470, 2023 May.
Article in English | MEDLINE | ID: mdl-35832012

ABSTRACT

Curcumin is a polyphenolic compound derived from the plant turmeric and the structural instability of which limits its further clinical applications. In this study, 11 curcumin analogs with more stable scaffold were prepared and evaluated. The results indicated that the optimal compound Y-11 exhibited the strongest antiproliferative activities against lung cancer cells including H460 and H1650. Further studies showed that Y-11 potentially inhibited hDHODH, induced cell cycle arrest and apoptosis as well as down-regulated crucial signal pathway protein expression in H1650 cells. In the conclusion, the newly designed curcumin analog Y-11 may be suitable for further development in lung cancer treatment.


Subject(s)
Antineoplastic Agents , Curcumin , Lung Neoplasms , Curcumin/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Cell Cycle Checkpoints , Apoptosis , Cell Proliferation
7.
Int J Mol Sci ; 24(11)2023 Jun 02.
Article in English | MEDLINE | ID: mdl-37298600

ABSTRACT

Breast Cancer (BC) is one of the most common and challenging cancers among females worldwide. Conventional treatments for oral cancer rely on the use of radiology and surgery accompanied by chemotherapy. Chemotherapy presents many side effects, and the cells often develop resistance to this chemotherapy. It will be urgent to adopt alternative or complementary treatment strategies that are new and more effective without these negative effects to improve the well-being of patients. A substantial number of epidemiological and experimental studies reported that many compounds are derived from natural products such as curcumin and their analogs, which have a great deal of beneficial anti-BC activity by inducing apoptosis, inhibiting cell proliferation, migration, and metastasis, modulating cancer-related pathways, and sensitizing cells to radiotherapy and chemotherapy. In the present study, we investigated the effect of the curcumin-analog PAC on DNA repair pathways in MCF-7 and MDA-MB-231 human breast-cancer cell lines. These pathways are crucial for genome maintenance and cancer prevention. MCF-7 and MDA-MB-231 cells were exposed to PAC at 10 µM. MTT and LDH assays were conducted to evaluate the effects of PAC on cell proliferation and cytotoxicity. Apoptosis was assessed in breast cancer cell lines using flow cytometry with annexin/Pi assay. The expression of proapoptotic and antiapoptotic genes was determined by RT-PCR to see if PAC is active in programming cell death. Additionally, DNA repair signaling pathways were analyzed by PCR arrays focusing on genes being related and confirmed by quantitative PCR. PAC significantly inhibited breast-cancer cell proliferation in a time-dependent manner, more on MDA-MB-231 triple-negative breast cancer cells. The flow cytometry results showed an increase in apoptotic activity. These data have been established by the gene expression and indicate that PAC-induced apoptosis by an increased Bax and decreased Bcl-2 expression. Moreover, PAC affected multiple genes involved in the DNA repair pathways occurring in both cell lines (MCF-7 and MDA-MB231). In addition, our results suggest that PAC upregulated more than twice 16 genes (ERCC1, ERCC2, PNKP, POLL, MPG, NEIL2, NTHL1, SMUG1, RAD51D, RAD54L, RFC1, TOP3A, XRCC3, XRCC6BP1, FEN1, and TREX1) in MDA-MB-231, 6 genes (ERCC1, LIG1, PNKP, UNG, MPG, and RAD54L) in MCF-7, and 4 genes (ERCC1, PNKP, MPG, and RAD54L) in the two cell lines. In silico analysis of gene-gene interaction shows that there are common genes between MCF-7 and MDA-MB-321 having direct and indirect effects, among them via coexpression, genetic interactions, pathways, predicted and physical interactions, and shared protein domains with predicted associated genes indicating they are more likely to be functionally related. Our data show that PAC increases involvement of multiple genes in a DNA repair pathway, this certainly can open a new perspective in breast-cancer treatment.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Curcumin , Triple Negative Breast Neoplasms , Female , Humans , Curcumin/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Apoptosis , Cell Proliferation , Gene Expression , DNA Repair , Antineoplastic Agents/pharmacology , Xeroderma Pigmentosum Group D Protein/genetics , Xeroderma Pigmentosum Group D Protein/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , DNA Repair Enzymes/genetics
8.
Inflammopharmacology ; 30(6): 2167-2179, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36261684

ABSTRACT

BACKGROUND: The pathogenesis of inflammatory bowel disease (IBD) remains unclear. C66, a derivative of curcumin, reportedly exerts anti-inflammatory, antifibrotic and anti-apoptotic effects by targeting the JNK pathway. However, the effect of C66 against IBD is not clear. In this study, we aimed to investigate the effect of C66 against IBD. METHODS: C57BL/6J mice were treated with 2.5% DSS for 7 days, and then administered water for 3 days to develop the IBD mouse model. A mouse intestinal epithelial cell line, MODE-K, stimulated by lipopolysaccharide (LPS) was used as the in vitro model. The therapeutic effects of C66 were evaluated and the pharmacological mechanisms were explored. RESULTS: Compared to the model group, C66 treatment significantly reduced colitis-associated damage, including a decrease in disease activity index (DAI), a higher body weight and longer colon. In addition, the infiltration of distal inflammatory cells, loss of crypt tissues, and destruction of epithelial cells were reduced in C66-treated group. In addition, C66 treatment reduced fibrotic areas and inflammatory responses in the colon tissues, leading to increased epithelial cell proliferation and decreased apoptosis in colon. Furthermore, C66 treatment decreased the levels of p-JNK and p-P65, indicating that C66 inhibits the activation of the JNK and NF-κB signaling pathways induced by DSS in colon tissues. Finally, in vitro data show that C66 inhibited LPS-induced inflammation and apoptosis in small intestinal epithelial cells.  CONCLUSIONS: The curcumin analog C66 exhibits its anti-inflammatory effect by inhibiting the DSS-induced activation of JNK/NF-κB signaling pathways. C66 may be a potential candidate for the treatment of IBD.


Subject(s)
Colitis , Curcumin , NF-kappa B , Animals , Mice , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Curcumin/analogs & derivatives , Curcumin/therapeutic use , Dextran Sulfate , Lipopolysaccharides , Mice, Inbred C57BL , NF-kappa B/metabolism
9.
Cancer Sci ; 112(2): 815-827, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33316116

ABSTRACT

Curcumin has a variety of anticancer properties, but low bioavailability prevents its use in chemotherapeutic applications. To address this problem, we tested the efficacy of the synthetic curcumin analog B14 in breast cancer cells and explored the mechanism by which B14 inhibits proliferation and metastasis of breast cancer cells. We used the breast cancer cell line MCF-7, MDA-MB-231 to study the anticancer effects of B14 and assessed cell viability, cell migration and invasion, cell cycle, and apoptosis, in addition, the antitumor effect of B14 in vivo was examined in mice bearing MDA-MB-231 cells. We found that, as the concentration of B14 increased, cell viability decreased in a dose-dependent manner. Compound B14 exerted the best antitumor activity and selectivity for MCF-7 and MDA-M-231 cells (IC50  = 8.84 µmol/L and 8.33 µmol/L, respectively), while its IC50 value for MCF-10A breast epithelial cells was 34.96 µmol/L. B14 has been shown to be a multi-targeted drug that alters the expression of cyclin D1, cyclin E1, and cyclin-dependent kinase 2 (CDK2), and ultimately induces G1 phase cell cycle arrest. At the same time, B14 activates the mitochondrial apoptosis pathway in breast cancer cells. Furthermore, B14 was more effective than curcumin in inhibiting cell migration, invasion, and colony formation. In tumor-bearing mice, analog B14 significantly reduced tumor growth and inhibited cell proliferation and angiogenesis. The pharmacokinetic test found that B14 was more stable than curcumin in vivo. Our data reveal the therapeutic potential of the curcumin analog B14 and the underlying mechanisms to fight breast cancer cells.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/pathology , Curcumin/analogs & derivatives , Curcumin/pharmacokinetics , Animals , Apoptosis/drug effects , Biological Availability , Cell Movement/drug effects , Cell Survival/drug effects , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays
10.
Bioorg Chem ; 104: 104273, 2020 11.
Article in English | MEDLINE | ID: mdl-32956875

ABSTRACT

ß-ionone, a cyclic terpenoid compound present in many fruits, has been showed a broad spectrum of biological activities. In this paper, we synthesized a panel of ß-ionone derivatives and tested their anti-proliferation activity on cancer cell by the MTT assay. The results showed that most of the ß-ionone derivatives were more active than ß-ionone and curcumin. Particularly, the ß-ionone derivatives (1a, 1d and 1g) with ortho-substituents on the aromatic ring exhibited much stronger cytotoxicity than their corresponding meta- and para-substituted compounds. Importantly, the cytotoxicity of the ß-ionone derivatives (1a, 1d and 1g) were relationship with their reactive oxygen species (ROS)-generation abilities, which could lead to the redox imbalance, lipid peroxidation, the loss of mitochondrial membrane potential (MMP), the activation of Bax and Caspase 3, followed by cell apoptosis. This work suggest that the "ortho effect", the ROS-generation ability and drawing fluorine atom into drugs may play a potent role in enhancing the anticancer activity of ß-ionone derivatives.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Norisoprenoids/pharmacology , Reactive Oxygen Species/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Norisoprenoids/chemical synthesis , Norisoprenoids/chemistry , Structure-Activity Relationship
11.
Int J Mol Sci ; 21(15)2020 Jul 30.
Article in English | MEDLINE | ID: mdl-32751716

ABSTRACT

Drugs for the treatment of Alzheimer's disease (AD) are in urgent demand due to the unmet need and the social burden associated with the disease. Curcumin has been historically considered as a beneficial product for anti-aging and AD. However, many efforts to develop curcumin for clinical use are hindered mainly due to its poor bioavailability. Recent development in drug delivery and structural design has resolved these issues. In this study, we identified a small molecule, TML-6, as a potential drug candidate for AD through screening a panel of curcumin derivatives using six biomarker platforms related to aging biology and AD pathogenesis. The structural modification of TML-6 is designed to improve the stability and metabolism of curcumin. Cell biological studies demonstrated that TML-6 could inhibit the synthesis of the ß-amyloid precursor protein and ß-amyloid (Aß), upregulate Apo E, suppress NF-κB and mTOR, and increase the activity of the anti-oxidative Nrf2 gene. In the 3x-Tg AD animal model, TML-6 treatment resulted in significant improvement in learning, suppression of the microglial activation marker Iba-1, and reduction in Aß in the brain. Although TML-6 exhibited a greater improvement in bioavailability as compared to curcumin, formulation optimization and toxicological studies are under development to assure its druggability. Taken together, TML-6 meets the current strategy to develop therapeutics for AD, targeting the combination of the Aß cascade and aging-related biology processes.


Subject(s)
Alzheimer Disease/drug therapy , Curcumin/pharmacology , Inflammation/drug therapy , NF-E2-Related Factor 2/genetics , Plaque, Amyloid/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Brain/pathology , Curcumin/analogs & derivatives , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Inflammation/genetics , Inflammation/pathology , Mice , Microglia/drug effects , Neuroprotective Agents/pharmacology , Plaque, Amyloid/genetics , Plaque, Amyloid/pathology
12.
Cancer Sci ; 110(2): 596-607, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30499149

ABSTRACT

Many prostate cancer patients develop resistance to treatment called castration-resistant prostate cancer (CRPC) which is the major cause of recurrence and death. In the present study, four cyclohexanone curcumin analogs were synthesized. Additionally, their anticancer progression activity on CRPC cell lines, PC3 and PLS10 cells, was examined. We first determined their anti-metastasis properties and found that 2,6-bis-(4-hydroxy-3-methoxy-benzylidene)-cyclohexanone (2A) and 2,6-bis-(3,4-dihydroxy-benzylidene)-cyclohexanone (2F) showed higher anti-invasion properties against CRPC cells than curcumin. Analog 2A inhibited both MMP-2 and MMP-9 secretions and activities, whereas analog 2F reduced only MMP activities. These findings suggest that the compounds may inhibit CRPC cell metastasis by decreased extracellular matrix degradation. Analog 2A, the most potent analog, was then subjected to an in vivo study. Similar to curcumin, analog 2A was detectable in the serum of mice at 30 and 60 minutes after i.p. injections. Analog 2A and curcumin (30 mg/kg bodyweight) showed a similar ability to reduce tumor area in lungs of mice that were i.v. injected with PLS10 cells. Additionally, analog 2A showed superior growth inhibitory effect on PLS10 cells than that of curcumin both in vitro and in vivo. The compound inhibited PLS10 cells growth by induction of G1 phase arrest and apoptosis in vitro. Interestingly, analog 2A significantly decreased tumor growth with downregulation of cell proliferation and angiogenesis in PLS10-bearing mice. Taken together, we could summarize that analog 2A showed promising activities in inhibiting CRPC progression both in vitro and in vivo.


Subject(s)
Curcumin/pharmacology , Cyclohexanones/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Drug Resistance, Neoplasm/drug effects , Humans , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/metabolism , PC-3 Cells , Prostatic Neoplasms, Castration-Resistant/metabolism , Xenograft Model Antitumor Assays/methods
13.
Acta Pharmacol Sin ; 40(11): 1412-1423, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31000770

ABSTRACT

Excessive and abnormal vessel growth plays a critical role in the pathogenesis of many diseases, such as cancer. Angiogenesis is one of the hallmarks of cancer growth, invasion, and metastasis. Discovery of novel antiangiogenic agents would provide new insights into the mechanisms of angiogenesis, as well as potential drugs for cancer treatment. In the present study, we investigated the antiangiogenic activity of a series of monocarbonyl analogs of curcumin synthesized previously in our lab. We found that curcumin analog A2 displayed the full potential to be developed as a novel antiangiogenic agent. Curcumin analog A2 at and above 20 µM dramatically inhibited the migration and tube formation of human umbilical vein endothelial cells (HUVECs) in vitro, new microvessels sprouting from the rat aortic rings ex vivo and newly formed microvessels in chicken chorioallantoic membranes (CAMs) and Matrigel plus in vivo. We further demonstrated that curcumin analog A2 exerted its antiangiogenic activity mainly through inducing endothelial cell death via elevating NADH/NADPH oxidase-derived ROS. Curcumin analog A2 at the antiangiogenic concentrations also triggered autophagy in HUVECs, but this process is neither a pre-requisite for toxicity, leading to the cell death nor a protective response against the toxicity of curcumin analog A2. In conclusion, we demonstrate for the first time the potent antiangiogenic activity of the monocarbonyl curcumin analog A2, which could serve as a promising potential therapeutic agent for the prevention and treatment angiogenesis-related diseases, such as cancer.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Curcumin/analogs & derivatives , Curcumin/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Reactive Oxygen Species/metabolism , Animals , Apoptosis/drug effects , Autophagy/drug effects , Chickens , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Necroptosis/drug effects , Rats, Sprague-Dawley , Sequestosome-1 Protein/metabolism
14.
Molecules ; 24(13)2019 Jul 06.
Article in English | MEDLINE | ID: mdl-31284561

ABSTRACT

Melanoma is the leading cause of skin-cancer related deaths in North America. Metastatic melanoma is difficult to treat and chemotherapies have limited success. Furthermore, chemotherapies lead to toxic side effects due to nonselective targeting of normal cells. Curcumin is a natural product of Curcuma longa (turmeric) and has been shown to possess anti-cancer activity. However, due to its poor bioavailability and stability, natural curcumin is not an effective cancer treatment. We tested synthetic analogs of curcumin that are more stable. One of these derivatives, Compound A, has shown significant anti-cancer efficacy in colon, leukemia, and triple-negative inflammatory breast cancer cells. However, the effects of Compound A against melanoma cells have not been studied before. In this study, for the first time, we demonstrated the efficacy of Compound A for the selective induction of apoptosis in melanoma cells and its interaction with tamoxifen, taxol, and cisplatin. We found that Compound A induced apoptosis selectively in human melanoma cells by increasing oxidative stress. The anti-cancer activity of Compound A was enhanced when combined with tamoxifen and the combination treatment did not result in significant toxicity to noncancerous cells. Additionally, Compound A did not interact negatively with the anti-cancer activity of taxol and cisplatin. These results indicate that Compound A could be developed as a selective and effective melanoma treatment either alone or in combination with other non-toxic agents like tamoxifen.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Curcumin/pharmacology , Drug Interactions , Antineoplastic Agents, Phytogenic/chemical synthesis , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Curcumin/analogs & derivatives , Curcumin/chemical synthesis , Dose-Response Relationship, Drug , Humans , Melanoma/drug therapy , Melanoma/metabolism , Melanoma/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Structure , Oxidative Stress/drug effects , Paclitaxel/pharmacology , Tamoxifen/pharmacology
15.
Cancer Sci ; 109(10): 3285-3293, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30024080

ABSTRACT

Tumor angiogenesis inhibition is one of the most potent strategies in cancer chemotherapy. From past clinical studies, inhibition of the vascular endothelial growth factor pathway successfully treats malignant tumors. However, vascular endothelial growth factor inhibitors alone cannot cure tumors. Moreover, resistance to small molecule inhibitors has also been reported. Herein, we show the antiangiogenic potential of a newly synthesized curcumin analog, GO-Y078, that possibly functions through inhibition of actin stress fiber formation, resulting in mobility inhibition; this mechanism is different from that of vascular endothelial growth factor inhibition. In addition, we examined the detailed mechanism of action of the antiangiogenesis potential of GO-Y078 using human umbilical venous epithelial cells resistant to angiogenesis inhibitors (HUVEC-R). GO-Y078 inhibited the growth and mobility of HUVEC-R at 0.75 µmol/L concentration. Expression analyses by microarray and RT-PCR showed that expressions of genes including that of fibronectin 1 were significantly suppressed. Among these genes, fibronectin 1 is abundantly expressed and, therefore, seems to be a good target for GO-Y078. In a knockdown experiment using Si-oligo of fibronectin 1 (FN1), FN1 expression was decreased to half of that in mock experiments as well as GO-Y078. Knockdown of FN1 resulted in the suppression of HUVEC-R growth at 24 hours after treatment. Fibronectin is a key molecule contributing to angiogenesis that could be inhibited by GO-Y078. Thus, resistance to vascular endothelial growth factor inhibition can be overcome using GO-Y078.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Curcumin/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Fibronectins/metabolism , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Angiogenesis Inhibitors/therapeutic use , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Curcumin/pharmacology , Curcumin/therapeutic use , Fibronectins/genetics , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/pathology , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/blood supply , Neovascularization, Pathologic/pathology , RNA, Small Interfering/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Xenograft Model Antitumor Assays , Xenopus laevis
16.
Int J Mol Sci ; 19(4)2018 Apr 11.
Article in English | MEDLINE | ID: mdl-29641445

ABSTRACT

Extensive research has been done in the search for innovative treatments against colon adenocarcinomas; however, the incidence rate of patients remains a major cause of cancer-related deaths in Malaysia. Natural bioactive compounds such as curcumin have been substantially studied as an alternative to anticancer drug therapies and have been surmised as a potent agent but, nevertheless, remain deficient due to its poor cellular uptake. Therefore, efforts now have shifted toward mimicking curcumin to synthesize novel compounds sharing similar effects. A synthetic analog, (Z)-3-hydroxy-1-(2-hydroxyphenyl)-3-phenylprop-2-ene-1-one (DK1), was recently synthesized and reported to confer improved bioavailability and selectivity toward human breast cancer cells. This study, therefore, aims to assess the anticancer mechanism of DK1 in relation to the induction of in vitro cell death in selected human colon cancer cell lines. Using the3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide(MTT) assay, the cytotoxicity of DK1 towards HT29 and SW620 cell lines were investigated. Acridine orange/propidium iodide (AO/PI) dual-staining assay and flow cytometry analyses (cell cycle analysis, Annexin/V-FITC and JC-1 assays) were incorporated to determine the mode of cell death. To further determine the mechanism of cell death, quantitative real-time polymerase chain reaction (qRT-PCR) and proteome profiling were conducted. Results from this study suggest that DK1 induced changes in cell morphology, leading to a decrease in cell viability and subsequent induction of apoptosis. DK1 treatment inhibited cell viability and proliferation 48 h post treatment with IC50 values of 7.5 ± 1.6 µM for HT29 cells and 14.5 ± 4.3 µM for SW620 cells, causing cell cycle arrest with increased accumulation of cell populations at the sub-G0/G1phaseof 74% and 23%, respectively. Flow cytometry analyses showed that DK1 treatment in cancer cells induced apoptosis, as indicated by DNA fragmentation and depolarization of the mitochondrial membrane. qRT-PCR results show significant upregulation in the expression of caspase-9 in both HT29 and SW620 cell lines, further supporting that cell death induction by DK1 is via an intrinsic pathway. These outcomes, therefore, demonstrate DK1 as a potential anticancer agent for colon adenocarcinoma due to its anti-apoptotic attributes.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma/metabolism , Colonic Neoplasms/metabolism , Curcumin/analogs & derivatives , Curcumin/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Curcumin/chemical synthesis , Curcumin/chemistry , HT29 Cells , Humans , Mitochondria/metabolism , Signal Transduction
17.
Molecules ; 23(1)2018 Jan 05.
Article in English | MEDLINE | ID: mdl-29303982

ABSTRACT

Osteosarcoma is one of the primary malignant bone tumors that confer low survival rates for patients even with intensive regime treatments. Therefore, discovery of novel anti-osteosarcoma drugs derived from natural products that are not harmful to the normal cells remains crucial. Curcumin is one of the natural substances that have been extensively studied due to its anti-cancer properties and is pharmacologically safe considering its ubiquitous consumption for centuries. However, curcumin suffers from a poor circulating bioavailability, which has led to the development of a chemically synthesized curcuminoid analog, namely (Z)-3-hydroxy-1-(2-hydroxyphenyl)-3-phenylprop-2-en-1-one (DK1). In this study, the cytotoxic effects of the curcumin analog DK1 was investigated in both U-2OS and MG-63 osteosarcoma cell lines using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and cell death was microscopically examined via acridine orange/propidium iodide (AO/PI) double staining. Flow cytometer analysis including Annexin V/Fluorescein isothiocyanate (FITC), cell cycle analysis and JC-1 were adapted to determine the mode of cell death. Subsequently in order to determine the mechanism of cell death, quantitative polymerase chain reaction (qPCR) and proteome profiling was carried out to measure the expression of several apoptotic-related genes and proteins. Results indicated that DK1 induced U-2 OS and MG-63 morphological changes and substantially reduced cell numbers through induction of apoptosis. Several apoptotic genes and proteins were steadily expressed after treatment with DK1; including caspase 3, caspase 9, and BAX, which indicated that apoptosis occurred through a mitochondria-dependent signaling pathway. In conclusion, DK1 could be considered as a potential candidate for an anti-osteosarcoma drug in the near future, contingent upon its ability to induce apoptosis in osteosarcoma cell lines.


Subject(s)
Antineoplastic Agents/pharmacology , Curcumin/analogs & derivatives , Curcumin/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Gene Expression/drug effects , Humans , Inhibitory Concentration 50 , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Signal Transduction/drug effects
18.
Int J Cancer ; 141(9): 1856-1866, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28646535

ABSTRACT

Advanced ovarian clear cell carcinoma (OCCC) carries a very poor prognosis in large part secondary to the extremely high rate of resistance to standard platinum and taxane chemotherapy. Signal transducer and activator of transcription 3(STAT3) expression and activation has been shown to regulate tumor progression in various human cancers, though has not been well studied in OCCC. Preliminary work in our lab has demonstrated constitutive activation of STAT3 (pSTAT3Tyr705 or pSTAT3727) in OCCC cell lines as well as human OCCC tumor tissue samples. Significantly, pSTAT3 is expressed in the absence of other forms of activated STAT (pSTAT1, 2, 6). Therefore, this work was planned to investigate the role of STAT3 and examine the efficacy of a novel anti-cancer compound -HO-3867, which is an inhibitor of STAT3, using known OCCC cell lines. Results demonstrate that treatment with HO-3867 decreased expression of pSTAT3 Tyr705 as well pSTAT3 Ser727, while total STAT3 remained constant. STAT3 overexpression increased the migration capability in OVTOKO cells in vitro and led to an increased tumor size when injected in vivo. The inhibitory effect of HO-3867 on cell proliferation and cell survival was accompanied by increased apoptosis, within 24 h post treatment. Treatment with HO-3867 resulted in a decrease in Bcl-2 and increase of cleavage of caspase 3, caspase 7, and PARP, confirming induction of apoptosis after treatment with HO-3867. In addition, HO-3867 significantly inhibited formation of human umbilical vein endothelial cells capillary-like structures and invasion at both 5 and 10 µM concentrations. STAT3 expression plays an important role in the spread of OCCC in vitro as well as in vivo. Thus, we can exploit the STAT3 pathway for targeted drug therapy. Inhibition of pSTAT3 using HO-3867in OCCC cell lines appears to be a promising therapy. This is of utmost importance given the poor response of OCCC to standard chemotherapy regimens.


Subject(s)
Adenocarcinoma, Clear Cell/drug therapy , Ovarian Neoplasms/drug therapy , Piperidones/administration & dosage , STAT3 Transcription Factor/genetics , Adenocarcinoma, Clear Cell/genetics , Adenocarcinoma, Clear Cell/pathology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
19.
Prostate ; 77(5): 489-504, 2017 04.
Article in English | MEDLINE | ID: mdl-27990666

ABSTRACT

BACKGROUND: The limited treatment option for recurrent prostate cancer and eventual resistant to conventional chemotherapy drugs has fueled continued interest in finding new anti-neoplastic agents. WZ35, a chemical analog of curcumin, had been demonstrated to have high chemical stability and potential anticancer effects in gastric cancer cells. The present study aimed to investigate the anti-prostate cancer effects of WZ35 in vitro and in vivo as well as the underlying mechanism. METHODS: Two prostate cancer cell lines RM-1 and DU145 were utilized to test the anti-cancer effects of WZ35 and the underlying mechanism. MTT assay was used to assess the cytotoxic effect of WZ35. Cell cycle distribution, apoptosis, alteration of ROS, and [Ca2+ ]i level were evaluated using flow cytometry. Western blotting assay was applied to measure the levels of proteins associated with apoptosis and cell cycle. Immunofluorescence staining and Electron micrographs were used to evaluate activation of mitochondrial apoptosis pathway. Tumor models in nude mice were induced by injection of RM-1 prostate cancer cells to test the in vivo anticancer action of WZ35. RESULTS: Our results showed that WZ35 treatment induced loss of cell viability, cell apoptosis, and G2/M cycle arrest in both RM-1 and DU145 cells, coupled with ROS overproduction, intracellular calcium surge, and activation of mitochondrial apoptosis pathway in RM-1 cells. Interestingly, all above changes induced by WZ35 were completely reversed by ROS blockage. In addition, prevention of [Ca2+ ]i elevation by BAPTA/AM also inhibited activation of mitochondrial apoptosis pathway induced by WZ35. In vivo studies, WZ35 treatment significantly inhibited RM-1 homograft tumor growth along with increased ROS accumulation, mitochondrial disruption, and cell apoptosis in tumor tissues. CONCLUSIONS: In conclusion, this work provides a novel anticancer candidate for the treatment of prostate cancer and demonstrated that increased ROS mediate the anti-cancer effects of WZ35 via activating mitochondrial apoptosis pathway. Importantly, this work also reveals that targeting ROS generation might be an effective strategy in human androgen-resistant prostate cancer treatment. Prostate 77:489-504, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/physiology , Intracellular Fluid/metabolism , Mitochondria/metabolism , Prostatic Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Humans , Intracellular Fluid/drug effects , Male , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondria/drug effects , Prostatic Neoplasms/drug therapy , Rats , Signal Transduction/drug effects , Signal Transduction/physiology , Treatment Outcome , Tumor Microenvironment/drug effects , Tumor Microenvironment/physiology , Xenograft Model Antitumor Assays/methods
20.
Mol Carcinog ; 56(7): 1765-1777, 2017 07.
Article in English | MEDLINE | ID: mdl-28218464

ABSTRACT

Lung cancer is the leading cause of cancer-related deaths. Curcumin is a well-known natural product with anticancer ability, however, its poor bioavailability and pharmacokinetic profiles have limited its application in anticancer therapy. Previously, we reported that L48H37, a novel analog of curcumin with higher bioavailability, ameliorated LPS-induced inflammation, but the anticancer effect of L48H37 is still unknown. In the present study, we have investigated the effects of L48H37 in human lung cancer cells. Our results show that L48H37 decreases lung cancer cell growth and colony formation. These alterations were mediated through induction of G2/M cell cycle arrest and apoptosis in lung cancer cells. After L48H37 treatment, ER stress-related proteins were increased, and the expression of p-STAT3 was decreased in a dose-dependent manner. L48H37 also induced the accumulation of ROS in lung cancer cells, and pretreatment with NAC could fully reverse L48H37-induced reactive oxygen species (ROS) increase. Blocking ROS was able to reverse L48H37-induced endoplasmic reticulum (ER) stress, cell cycle arrest, and apoptosis. Finally, we show that L48H37 inhibits the growth of lung cancer xenografts without exhibiting toxicity. Treatment of mice bearing human lung cancer xenografts with L48H37 was also associated with indices of ER stress activation. In summary, our results provide evidence for a novel anti-tumor candidate for the treatment of lung cancer.


Subject(s)
Apoptosis/drug effects , Curcumin/analogs & derivatives , Endoplasmic Reticulum Stress/drug effects , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Curcumin/pharmacology , Female , Humans , Lung Neoplasms/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL