1
|
Song B, Kim D, Ho JN, Le VH, Lee S. Crizotinib Inhibits Viability, Migration, and Invasion by Suppressing the c-Met/ PI3K/ Akt Pathway in the Three-Dimensional Bladder Cancer Spheroid Model. Curr Oncol 2025; 32:236. [PMID: 40277792 PMCID: PMC12025888 DOI: 10.3390/curroncol32040236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/01/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
We aimed to evaluate the therapeutic potential of crizotinib, a broad-spectrum tyrosine kinase inhibitor against bladder cancer (BC) cells, based on a three-dimensional (3D) cell culture system. After proliferating cell masses (spheroids) using T24 cisplatin-naïve and T24R2 cisplatin-resistant human BC cell lines, the spheroids were exposed to various crizotinib concentrations in order to derive an ideal crizotinib concentration to suppress cell survival, migration, and invasion. Crizotinib suppressed cell proliferation, migration, and invasion in both T24 and T24R2 BC cell lines under a 3D spheroid model, which was more appropriate than the conventional two-dimensional cell culture model. Real-time quantitative polymerase chain reaction analysis revealed a reduced expression of E-cadherin and an enhanced expression of vimentin, suggesting EMT suppression and the subsequent suppression of tumor aggressiveness following crizotinib administration. Meanwhile, the expressions of apoptosis-related genes increased. Western blot analysis revealed that the expression levels of phosphorylated mesenchymal-epithelial transition factor (c-Met) and phosphorylated Akt decreased following crizotinib administration, suggesting that the antitumor effect of crizotinib can be associated with the inhibition of the phosphorylated activation of the c-Met/PI3K/Akt pathway. Crizotinib showed a potential antitumor effect on both cisplatin-naïve and cisplatin-resistant human BC cells, likely through c-Met-induced PI3K/Akt pathway inhibition.
Collapse
Affiliation(s)
- Byeongdo Song
- Department of Urology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri-si 11923, Gyeonggi-do, Republic of Korea;
| | - Danhyo Kim
- Department of Urology, Seoul National University Bundang Hospital, 166, Gumi-ro, Bundang-gu, Seongnam-si 13620, Gyunggi-do, Republic of Korea; (D.K.); (J.-N.H.); (V.-H.L.)
| | - Jin-Nyoung Ho
- Department of Urology, Seoul National University Bundang Hospital, 166, Gumi-ro, Bundang-gu, Seongnam-si 13620, Gyunggi-do, Republic of Korea; (D.K.); (J.-N.H.); (V.-H.L.)
| | - Van-Hung Le
- Department of Urology, Seoul National University Bundang Hospital, 166, Gumi-ro, Bundang-gu, Seongnam-si 13620, Gyunggi-do, Republic of Korea; (D.K.); (J.-N.H.); (V.-H.L.)
- Department of Urology, Vinmec International Hospital, 458 P. Minh Khai, Hanoi 100000, Vietnam
| | - Sangchul Lee
- Department of Urology, Seoul National University Bundang Hospital, 166, Gumi-ro, Bundang-gu, Seongnam-si 13620, Gyunggi-do, Republic of Korea; (D.K.); (J.-N.H.); (V.-H.L.)
- Department of Urology, Seoul National University College of Medicine, 71, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| |
Collapse
|
2
|
Lee JH, Lim JY, Jeon YD, Kim DK, Lee DH. Methanol Extract of Pueraria lobata (Willd.) Root and Its Active Ingredient, Puerarin, Induce Apoptosis in HeLa Cells and Attenuates Bacterial Vaginosis in Gardnerella vaginalis-Infected Mice. Int J Mol Sci 2025; 26:1342. [PMID: 39941110 PMCID: PMC11818357 DOI: 10.3390/ijms26031342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
Pueraria lobata (Willd.) has been used as food since ancient times, and its roots have been used mainly as a traditional herbal medicine to treat various diseases in East Asia. Puerarin is one of the major active ingredients in the roots of P. lobata. The purpose of this study was to examine the effects of the methanol extract of P. lobata roots (PRME) and puerarin on apoptosis in cervical cancer and inflammation-relieving effects in vaginitis. First, we prepared the PRME and confirmed the puerarin content of PRME through HPLC analysis. We performed a TUNEL assay, Hoechst 33342 staining, and western blotting using HeLa cells, a human cervical cancer cell line. Both the PRME and puerarin exhibited antiproliferative effects in HeLa cells by inducing apoptosis through the activation of the extrinsic death receptor and intrinsic mitochondrial pathways, thereby demonstrating their anticancer efficacy against human cervical cancer. Next, a mouse model of vaginitis induced by Gardnerella vaginalis (GV) infection was established by inoculating C57BL/6 mice with β-estradiol-3-benzoate and GV (1 × 108 CFU). Histological analysis and PCR confirmed that the administration of PRME or puerarin to GV-infected mice alleviated reproductive tract vaginitis symptoms. Additionally, we confirmed that PRME or puerarin treatment decreased myeloperoxidase activity and reduced inflammation by regulating cytokines through the secretion of inflammatory mediators in mouse vaginal tissue. These results demonstrate that PRME and puerarin can be used as potential adjuvants or therapeutic agents with anticancer and anti-inflammatory properties to inhibit the progression of human cervical cancer and alleviate vaginitis.
Collapse
Affiliation(s)
- Ji-Hyun Lee
- Department of Immunology, Medical School, Jeonbuk National University, Jeonju 54907, Republic of Korea; (J.-H.L.); (J.-Y.L.)
| | - Ji-Ye Lim
- Department of Immunology, Medical School, Jeonbuk National University, Jeonju 54907, Republic of Korea; (J.-H.L.); (J.-Y.L.)
| | - Yong-Deok Jeon
- Department of Korean Pharmacy, Woosuk University, Wanju 55338, Republic of Korea;
| | - Dae-Ki Kim
- Department of Immunology, Medical School, Jeonbuk National University, Jeonju 54907, Republic of Korea; (J.-H.L.); (J.-Y.L.)
| | - Dong-Hyun Lee
- Department of Obstetrics and Gynecology, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
| |
Collapse
|
3
|
Yang Q, Meng D, Zhang Q, Wang J. Advances in research on the anti-tumor mechanism of Astragalus polysaccharides. Front Oncol 2024; 14:1334915. [PMID: 38515577 PMCID: PMC10955345 DOI: 10.3389/fonc.2024.1334915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 02/22/2024] [Indexed: 03/23/2024] Open
Abstract
The dry root of the soybean plant Astragalus membranaceus (Fisch) Bge. var. mongholicus (Bge) Hsiao or A. membranaceus (Fisch) Bge, Astragali Radix (AR) has a long medicinal history. Astragalus polysaccharide (APS), the natural macromolecule that exhibits immune regulatory, anti-inflammatory, anti-tumor, and other pharmacological activities, is an important active ingredient extracted from AR. Recently, APS has been increasingly used in cancer therapy owing to its anti-tumor ability as it prevents the progression of prostate, liver, cervical, ovarian, and non-small-cell lung cancer by suppressing tumor cell growth and invasion and enhancing apoptosis. In addition, APS enhances the sensitivity of tumors to antineoplastic agents and improves the body's immunity. This macromolecule has prospects for broad application in tumor therapy through various pathways. In this article, we present the latest progress in the research on the anti-tumor effects of APS and its underlying mechanisms, aiming to provide novel theoretical support and reference for its use in cancer therapy.
Collapse
Affiliation(s)
| | | | - Qinyuan Zhang
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jin Wang
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
4
|
Fawzi M, Bimoussa A, Laamari Y, Oussidi AN, Oubella A, Ketatni EM, Saadi M, Ammari LE, Morjani H, Ait Itto MY, Auhmani A. New (S)-verbenone-isoxazoline-1,3,4-thiadiazole hybrids: synthesis, anticancer activity and apoptosis-inducing effect. Future Med Chem 2023; 15:1603-1619. [PMID: 37772541 DOI: 10.4155/fmc-2023-0173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023] Open
Abstract
Background: This study aimed to develop novel isoxazoline-1,3,4-thiadiazole hybrids from (S)-verbenone for potential anticancer treatment, particularly focusing on cytotoxic and apoptotic effects in hormone-sensitive MCF-7 and triple-negative MDA-MB-231 breast cancer cells. Methods & results: (S)-verbenone was used to synthesize hybrids through 1,3-dipolar cycloaddition, followed by thorough characterization. The compounds were screened across cancer cell lines, showing significant anticancer effects. Compound 8b notably induced apoptosis via the caspase-3/7 pathway and cell cycle arrest, displaying noteworthy cytotoxicity against MCF-7 and MDA-MB-231 cells. Conclusion: These findings underscore the potential of (S)-verbenone isoxazoline-1,3,4-thiadiazole derivatives for breast cancer therapy due to their remarkable apoptotic activity. This study highlights a promising avenue for advancing breast cancer treatment using these derivatives, founded on (S)-verbenone, showcasing their distinct potential for inducing apoptosis.
Collapse
Affiliation(s)
- Mourad Fawzi
- Laboratory of Molecular Chemistry, Unit of Organic Synthesis & Molecular Physicochemistry, Department of Chemistry, Faculty of Sciences Semlalia, PO Box 2390, Marrakech, 40001, Morocco
| | - Abdoullah Bimoussa
- Laboratory of Molecular Chemistry, Unit of Organic Synthesis & Molecular Physicochemistry, Department of Chemistry, Faculty of Sciences Semlalia, PO Box 2390, Marrakech, 40001, Morocco
| | - Yassine Laamari
- Laboratory of Molecular Chemistry, Unit of Organic Synthesis & Molecular Physicochemistry, Department of Chemistry, Faculty of Sciences Semlalia, PO Box 2390, Marrakech, 40001, Morocco
| | - Abdellah N'ait Oussidi
- Laboratory of Molecular Chemistry, Unit of Organic Synthesis & Molecular Physicochemistry, Department of Chemistry, Faculty of Sciences Semlalia, PO Box 2390, Marrakech, 40001, Morocco
| | - Ali Oubella
- Laboratory of Organic & Physical Chemistry, Applied Bioorganic Chemistry Team, Faculty of Sciences, IBNOU ZOHR University, Agadir, 80000, Morocco
| | - El Mostafa Ketatni
- Laboratory of Molecular Chemistry, Materials & Catalysis, Faculty of Sciences, & Technics, Sultan Moulay Slimane University, Beni-Mellal, BP 523, 23000, Morocco
| | - Mohamed Saadi
- Laboratoire de Chimie Appliquée des Matériaux, Centre des Sciences des Matériaux, Faculty of Sciences, Mohammed V University in Rabat, Avenue Ibn Batouta, PO Box 1014, Rabat, Morocco
| | - Lahcen El Ammari
- Laboratoire de Chimie Appliquée des Matériaux, Centre des Sciences des Matériaux, Faculty of Sciences, Mohammed V University in Rabat, Avenue Ibn Batouta, PO Box 1014, Rabat, Morocco
| | - Hamid Morjani
- Unité BioSpecT, EA7506, SFR CAP-Santé, UFR de Pharmacie, Université de Reims Champagne-Ardenne, 51100, France
| | - Moulay Youssef Ait Itto
- Laboratory of Molecular Chemistry, Unit of Organic Synthesis & Molecular Physicochemistry, Department of Chemistry, Faculty of Sciences Semlalia, PO Box 2390, Marrakech, 40001, Morocco
| | - Aziz Auhmani
- Laboratory of Molecular Chemistry, Unit of Organic Synthesis & Molecular Physicochemistry, Department of Chemistry, Faculty of Sciences Semlalia, PO Box 2390, Marrakech, 40001, Morocco
| |
Collapse
|
5
|
Wang H, Liu J, Wang M, Yang C, Wang G, Hu T. The adverse effect of anticancer drug toremifene on vascular smooth muscle cells is an important aspect of its tumor growth inhibition. J Cancer Res Clin Oncol 2023; 149:7837-7848. [PMID: 37036506 DOI: 10.1007/s00432-023-04744-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/03/2023] [Indexed: 04/11/2023]
Abstract
PURPOSE Toremifene (TOR) is widely used as an antineoplastic drug and has an inhibitory effect on angiogenesis in mesenteric desmoid tumors and vascular intracranial solitary fibrous tumors. However, no study has investigated the direct effect of TOR on vascular cells. This study aimed at exploring the effect of TOR on the behaviors of vascular smooth muscle cells (VSMCs). METHODS Human aortic umbilical vascular smooth muscle cells (HAVSMCs) were treated by TOR. Cell morphology, migration, adhesion, and proliferation assay were investigated. The cell cycle, apoptosis, mitochondrial membrane potential, and reactive oxygen species were assessed using flow cytometry. Caspase-3 and 9 activities were assayed using Caspase-3 and Caspase-9 Activity Assay kits, respectively. Immunofluorescence and Western blot assays were carried out to characterize protein expressions of PCNA, p53, and Rho/ROCK signaling pathway. RESULTS TOR damaged cytoskeleton, inhibited VSMC proliferation, migration, and adhesion, and induced abnormal cell morphology and apoptosis. The antiproliferative activity of TOR was associated with the induction of G0/G1 phase arrest, blocking the cell cycle. TOR disrupted intracellular reactive oxygen species and mitochondrial membrane potential, and enhanced p53 expression and the activities of caspase-3 and caspase-9. Thus, TOR-induced apoptosis by the mitochondrial signaling pathway. Additionally, TOR induced decreased Rho, ROCK, MLC, and pMLC proteins. Collectively, TOR may affect multiple behaviors of VSMCs by damaging cytoskeleton through the Rho/ROCK pathway. CONCLUSION The adverse effect of TOR on VSMCs could be considered as an important aspect of tumor growth inhibition.
Collapse
Affiliation(s)
- Huiyun Wang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Juan Liu
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Mingxing Wang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Chun Yang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Guixue Wang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Tingzhang Hu
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
6
|
Joglekar I, Clark AC. Sequential Unfolding Mechanisms of Monomeric Caspases. Biochemistry 2023; 62:1878-1889. [PMID: 37337671 DOI: 10.1021/acs.biochem.3c00004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
Caspases are evolutionarily conserved cysteinyl proteases that are integral in cell development and apoptosis. All apoptotic caspases evolved from a common ancestor into two distinct subfamilies with either monomeric (initiators) or dimeric (effectors) oligomeric states. The regulation of apoptosis is influenced by the activation mechanism of the two subfamilies, but the evolution of the well-conserved caspase-hemoglobinase fold into the two subfamilies is not well understood. We examined the folding landscape of monomeric caspases from two coral species over a broad pH range of 3-10.5. On an evolutionary timescale, the two coral caspases diverged from each other approximately 300 million years ago, and they diverged from human caspases about 600 million years ago. Our results indicate that both proteins have overall high stability, ∼15 kcal mol-1, near the physiological pH range (pH 6-8) and unfold via two partially folded intermediates, I1 and I2*, that are in equilibrium with the native and the unfolded state. Like the dimeric caspases, the monomeric coral caspases undergo a pH-dependent conformational change resulting from the titration of an evolutionarily conserved site. Data from molecular dynamics simulations paired with limited proteolysis and MALDI-TOF mass spectrometry show that the small subunit of the monomeric caspases is unstable and unfolds prior to the large subunit. Overall, the data suggest that all caspases share a conserved folding landscape, that a conserved allosteric site can be fine-tuned for species-specific regulation, and that the subfamily of stable dimers may have evolved to stabilize the small subunit.
Collapse
Affiliation(s)
- Isha Joglekar
- Department of Biology, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - A Clay Clark
- Department of Biology, University of Texas at Arlington, Arlington, Texas 76019, United States
| |
Collapse
|
7
|
Li M, Zhang R, Ge Q, Yue L, Ma D, Khattab F, Xie W, Cui Y, Gilon P, Zhao X, Li X, Cheng R. Chemerin as an Inducer of β Cell Proliferation Mediates Mitochondrial Homeostasis and Promotes β Cell Mass Expansion. Int J Mol Sci 2023; 24:ijms24119136. [PMID: 37298086 DOI: 10.3390/ijms24119136] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
Loss of the β cell population is a crucial feature of type 2 diabetes. Restoring the β cell mass by stimulating β cell proliferation and preventing its apoptosis was proposed as a therapeutic approach to treating diabetes. Therefore, researchers have been increasingly interested in identifying exogenous factors that can stimulate β cell proliferation in situ and in vitro. Adipokine chemerin, which is secreted from adipose tissue and the liver, has been identified as a chemokine that plays a critical role in the regulation of metabolism. In this study, we demonstrate that chemerin as a circulating adipokine promotes β cell proliferation in vivo and in vitro. Chemerin serum levels and the expression of the main receptors within islets are highly regulated under a variety of challenging conditions, including obesity and type 2 diabetes. As compared to their littermates, mice overexpressing chemerin had a larger islet area and increased β cell mass with both a normal and high-fat diet. Moreover, in chemerin-overexpressed mice, we observed improved mitochondrial homeostasis and increased insulin synthesis. In summary, our findings confirm the potential role of chemerin as an inducer of β cell proliferation, and they provide novel insights into the helpful strategy to expand β cell population.
Collapse
Affiliation(s)
- Min Li
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Ruifan Zhang
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Qian Ge
- The First Clinical College, Chongqing Medical University, Chongqing 400016, China
| | - Lingzhi Yue
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Dan Ma
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Firas Khattab
- Pôle d'Endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Wenhua Xie
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yewei Cui
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Patrick Gilon
- Pôle d'Endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Xueya Zhao
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xi Li
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Rui Cheng
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
8
|
Lu Q, Huang H, Wang X, Luo L, Xia H, Zhang L, Xu J, Huang Y, Luo X, Luo J. Echinatin inhibits the growth and metastasis of human osteosarcoma cells through Wnt/β-catenin and p38 signaling pathways. Pharmacol Res 2023; 191:106760. [PMID: 37023991 DOI: 10.1016/j.phrs.2023.106760] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/01/2023] [Accepted: 04/04/2023] [Indexed: 04/08/2023]
Abstract
Osteosarcoma (OS) is a highly aggressive malignant bone tumor that mainly occurs in adolescents. At present, chemotherapy is the most commonly used method in clinical practice to treat OS. However, due to drug resistance, toxicity and long-term side effects, chemotherapy can't always provide sufficient benefits for OS patients, especially those with metastasis and recurrence. Natural products have long been an excellent source of anti-tumor drug development. In the current study, we evaluated the anti-OS activity of Echinatin (Ecn), a natural active component from the roots and rhizomes of licorice, and explored the possible mechanism. We found that Ecn inhibited the proliferation of human OS cells and blocked cell cycle at S phase. In addition, Ecn suppressed the migration and invasion, while induced the apoptosis of human OS cells. However, Ecn had less cytotoxicity against normal cells. Moreover, Ecn inhibited the xenograft tumor growth of OS cells in vivo. Mechanistically, Ecn inactivated Wnt/β-catenin signaling pathway while activated p38 signaling pathway. β-catenin over-expression and the p38 inhibitor SB203580 both attenuated the inhibitory effect of Ecn on OS cells. Notably, we demonstrated that Ecn exhibited synergistic inhibitory effect with cisplatin (DDP) on OS cells in vitro and in vivo. Therefore, our results suggest that Ecn may exert anti-OS effects at least partly through regulating Wnt/β-catenin and p38 signaling pathways. Most meaningfully, the results obtained suggest a potential strategy to improve the DDP-induced tumor-killing effect on OS cells by combining with Ecn.
Collapse
Affiliation(s)
- Qiuping Lu
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 40016, China
| | - Huakun Huang
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 40016, China
| | - Xiaoxuan Wang
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 40016, China
| | - Lijuan Luo
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 40016, China
| | - Haichao Xia
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 40016, China
| | - Lulu Zhang
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 40016, China
| | - Jingtao Xu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 40016, China
| | - Yanran Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 40016, China
| | - Xiaoji Luo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 40016, China
| | - Jinyong Luo
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 40016, China.
| |
Collapse
|
9
|
Joglekar I, Clark AC. Sequential unfolding mechanisms of monomeric caspases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.04.522771. [PMID: 36711547 PMCID: PMC9881926 DOI: 10.1101/2023.01.04.522771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Caspases are evolutionarily conserved cysteinyl proteases that are integral in cell development and apoptosis. All apoptotic caspases evolved from a common ancestor into two distinct subfamilies with either monomeric (initiators) or dimeric (effectors) oligomeric states. The regulation of apoptosis is influenced by the activation mechanism of the two subfamilies, but the evolution of the well-conserved caspase-hemoglobinase fold into the two subfamilies is not well understood. We examined the folding landscape of monomeric caspases from two coral species over a broad pH range of 3 to 10.5. On an evolutionary timescale, the two coral caspases diverged from each other approximately 300 million years ago, and they diverged from human caspases about 600 million years ago. Our results indicate that both proteins have overall high stability, ∼ 15 kcal mol -1 near the physiological pH range (pH 6 to pH 8), and unfold via two partially folded intermediates, I 1 and I 2 , that are in equilibrium with the native and the unfolded state. Like the dimeric caspases, the monomeric coral caspases undergo a pH-dependent conformational change resulting from the titration of an evolutionarily conserved site. Data from molecular dynamics simulations paired with limited proteolysis and MALDI-TOF mass spectrometry show that the small subunit of the monomeric caspases is unstable and unfolds prior to the large subunit. Overall, the data suggest that all caspases share a conserved folding landscape, that a conserved allosteric site can be fine-tuned for species-specific regulation, and that the subfamily of stable dimers may have evolved to stabilize the small subunit.
Collapse
Affiliation(s)
- Isha Joglekar
- Department of Biology, University of Texas at Arlington, Arlington, Texas, 76019
| | - A. Clay Clark
- Department of Biology, University of Texas at Arlington, Arlington, Texas, 76019,Corresponding author: A. Clay Clark,
| |
Collapse
|
10
|
Ayoup MS, Mansour AF, Abdel-Hamid H, Abu-Serie MM, Mohyeldin SM, Teleb M. Nature-inspired new isoindole-based Passerini adducts as efficient tumor-selective apoptotic inducers via caspase-3/7 activation. Eur J Med Chem 2023; 245:114865. [DOI: 10.1016/j.ejmech.2022.114865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/06/2022] [Accepted: 10/18/2022] [Indexed: 11/29/2022]
|
11
|
Structure optimization of new tumor-selective Passerini α-acyloxy carboxamides as Caspase-3/7 activators. Sci Rep 2022; 12:22390. [PMID: 36575196 PMCID: PMC9794698 DOI: 10.1038/s41598-022-26469-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/15/2022] [Indexed: 12/28/2022] Open
Abstract
Selective elimination of tumors has always been the mainstay of oncology research. The on-going research underlying the cellular apoptotic mechanisms reveal caspases activation, especially the key effector caspase-3, as a personalized tumor-selective therapeutic strategy. Our continued research protocol has exploited new optimized Passerini α-acyloxy carboxamides as efficient apoptotic inducers via caspase-3/7 dependent mechanism with highly selective anticancer profiles. The adopted design rationale relied on excluding structural alerts of previous leads, while merging various pharmacophoric motifs of natural and synthetic caspase activators via optimized one-pot Passerini reaction conditions. The prepared compounds resulting from Passerini reaction were screened for their cytotoxic activities against colorectal Caco-2 and liver HepG-2 cancer cells compared to normal fibroblasts utilizing MTT assay. Notably, all compounds exhibited promising low-range submicromolar IC50 against the studied cancer cell lines, with outstanding tumor selectivity (SI values up to 266). Hence, they were superior to 5-fluorouracil. Notably, 7a, 7g, and 7j conferred the highest potencies against Caco-2 and HepG-2 cells and were selected for further mechanistic studies. Caspas-3/7 activation assay of the hit compounds and flow cytometric analysis of the treated apoptotic cancer cells demonstrated their significant caspase activation potential (up to 4.2 folds) and apoptotic induction capacities (up to 58.7%). Further assessment of Bcl2 expression was performed being a physiological caspase-3 substrate. Herein, the three studied Passerini adducts were able to downregulate Bcl2 in the treated Caco-2 cells. Importantly, the mechanistic studies results of the three hits echoed their preliminary MTT antiproliferative potencies data highlighting their caspase-3 dependent apoptotic induction. Finally, the in silico predicted physicochemical and pharmacokinetic profiles, as well as ligand efficiency metrics were drug-like.
Collapse
|
12
|
Isolation of Potential Compound from the Leaves of Elytraria acaulis and Evaluating Its Therapeutic Properties Using In Vitro Studies Against Ovarian Cancer. Appl Biochem Biotechnol 2022; 194:5607-5626. [PMID: 35796948 DOI: 10.1007/s12010-022-04048-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2022] [Indexed: 11/02/2022]
Abstract
The present study was designed to isolate a potential compound from the extracts of Elytraria acaulis (E. acaulis) for ovarian cancer. n-Hexane, ethyl acetate, chloroform, acetone and methanol extract were taken using the Soxhlet method. Thin layer, column chromatography, NMR and MASS studies were done for the isolation and structural characterization of the compound. Finally, the novel compound (Z)-3-(2-methyl-3-oxoprop-1-en-1-yl) phenyl heptanoate was identified. MTT assay, cell morphology and cell cycle analysis were done to evaluate the anticancer property of the compound. In the MTT assay, the percentage of the cell viability treated with the isolated compound was decreased while increasing the concentration of the compound. Cancer cells treated with the isolated compound showed distinct morphological changes when compared to the control untreated cells. In the cell cycle analysis, the isolated compound induced a significant increase in the percentage of cells in G0/G1 phase and a decrease in the percentage of cells in the S phase and G2-M phase of the PA 1 cell lines. The cell cycle arrest induced by the isolated compound may account for its antiproliferative capacity. Hence, the novel compound isolated from E. acaulis can be a potent candidate in the designing of anticancer drugs.
Collapse
|
13
|
Soonnarong R, Putra ID, Sriratanasak N, Sritularak B, Chanvorachote P. Artonin F Induces the Ubiquitin-Proteasomal Degradation of c-Met and Decreases Akt-mTOR Signaling. Pharmaceuticals (Basel) 2022; 15:ph15050633. [PMID: 35631459 PMCID: PMC9145792 DOI: 10.3390/ph15050633] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 12/04/2022] Open
Abstract
Targeted therapies that selectively inhibit certain molecules in cancer cells have been considered promising for cancer treatment. In lung cancer, evidence has suggested that mesenchymal-epithelial transition factor (c-Met) oncoprotein drives cancer progression through its signaling transduction pathway. In this paper, we report the downregulation of c-Met by artonin F, a flavonoid isolated from Artocarpus gomezianus. Artonin F was found to be dominantly toxic to lung cancer cells by mediating apoptosis. With regard to its mechanism of action, artonin F downregulated c-Met expression, consequently suppressed the phosphatidylinositol-3 kinase/Akt/mammalian target of rapamycin signaling, increased Bax expression, decreased Bcl-2 expression, and activated caspase-3. The depletion of c-Met was mediated by ubiquitin-proteasomal degradation following co-treatment with artonin F, with the proteasome inhibitor MG132 reversing its c-Met-targeting effect. The immunoprecipitation analysis revealed that artonin F significantly promoted the formation of the c-Met–ubiquitin complex. Given that ubiquitin-specific protease 8 (USP8) prevents c-Met degradation by deubiquitination, we performed a preliminary in silico molecular docking and observed that artonin F blocked the catalytic site of USP8. In addition, artonin F interacted with the catalytic residues of palmitoylating enzymes. By acting as a competitive inhibitor, artonin F could reduce the degree of palmitoylation of c-Met, which affected its stability and activity. In conclusion, c-Met is critical for cancer cell survival and the failure of chemotherapeutic regimens. This novel information on the c-Met downregulating effect of artonin F will be beneficial for the development of efficient anticancer strategies or targeted therapies.
Collapse
Affiliation(s)
- Rapeepun Soonnarong
- Interdisciplinary Program of Pharmacology Graduate School, Chulalongkorn University, Bangkok 10330, Thailand;
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (I.D.P.); (N.S.)
| | - Ismail Dwi Putra
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (I.D.P.); (N.S.)
- Pharmaceutical Sciences and Technology Graduate Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Nicharat Sriratanasak
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (I.D.P.); (N.S.)
- Departments of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Bangkok 10330, Thailand
| | - Boonchoo Sritularak
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Pithi Chanvorachote
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (I.D.P.); (N.S.)
- Departments of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Bangkok 10330, Thailand
- Correspondence: ; Tel.: +662-218-8344
| |
Collapse
|
14
|
Barathan M, Zulpa AK, Mee Hoong S, Vellasamy KM, Vadivelu J. Synergistic effect of hyperforin and paclitaxel on growth inhibition, apoptotic mediator activation in MCF-7 human breast cancer cells. JOURNAL OF TAIBAH UNIVERSITY FOR SCIENCE 2021. [DOI: 10.1080/16583655.2021.2010910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Muttiah Barathan
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Ahmad Khusairy Zulpa
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - See Mee Hoong
- Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Kumutha Malar Vellasamy
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Jamuna Vadivelu
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
15
|
Daneshforouz A, Nazemi S, Gholami O, Kafami M, Amin B. The cytotoxicity and apoptotic effects of verbascoside on breast cancer 4T1 cell line. BMC Pharmacol Toxicol 2021; 22:72. [PMID: 34844644 PMCID: PMC8628474 DOI: 10.1186/s40360-021-00540-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 11/22/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Despite significant advancements in breast cancer therapy, novel drugs with lower side effects are still being demanded. In this regard, we investigated the anti-cancer features of verbascoside in 4 T1 mouse mammary tumor cell. METHODS First, MTT assay was performed with various concentrations (ranging between 5 to 200 μM) of verbascoside and IC50 was calculated. Then the expression of Bax, Bcl-2, and caspase-3 was evaluated in treated 4 T1 cells. In addition, we investigated the expression of TLR4, MyD88, and NF-κB to ascertain the underlying mechanism of the anti-proliferative feature of verbascoside. Also, flow cytometry followed by double PI and Annexin V was conducted to confirm the apoptosis-inducing effect of verbascoside. RESULTS Our results from MTT assay showed verbascoside inhibits proliferation of 4 T1 cancer cells (IC50 117 μM) while is safe for normal HEK293T cells. By qRT-PCR, we observed that verbascoside treatment (100, 117 and, 130 μM) increases the expression of caspase-3 and Bax while reduces the expression of Bcl-2. Also, verbascoside (100, 117 and, 130 μM) increased the expression of TLR4 only at 130 μM dose and the expression of MyD88 whereas reduced the expression of NF-κB at mRNA level. Flow cytometry analysis also confirmed verbascoside induces apoptosis in 4 T1 cells at 117 μM. CONCLUSION Taken together, our data showed verbascoside is a safe natural compound for normal cells while has apoptosis-inducing feature through TLR4 axis on 4 T1 cells.
Collapse
Affiliation(s)
- Atena Daneshforouz
- Student Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Samad Nazemi
- Cellular and Molecular Research Center, Department of Physiology and Pharmacology, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Omid Gholami
- Cellular and Molecular Research Center, Department of Physiology and Pharmacology, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Marzieh Kafami
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran. .,Department of Physiology and Pharmacology, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran.
| | - Bahareh Amin
- Cellular and Molecular Research Center, Department of Physiology and Pharmacology, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran.
| |
Collapse
|
16
|
Antitumor and apoptotic effects of new-generation platinum compounds on human leukemia cell lines HL-60 and K562. Biologia (Bratisl) 2021. [DOI: 10.1007/s11756-021-00930-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
17
|
Alsaif NA, Taghour MS, Alanazi MM, Obaidullah AJ, Alanazi WA, Alasmari A, Albassam H, Dahab MA, Mahdy HA. Identification of new [1,2,4]triazolo[4,3-a]quinoxalines as potent VEGFR-2 tyrosine kinase inhibitors: Design, synthesis, anticancer evaluation, and in silico studies. Bioorg Med Chem 2021; 46:116384. [PMID: 34479065 DOI: 10.1016/j.bmc.2021.116384] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/12/2021] [Accepted: 08/23/2021] [Indexed: 11/16/2022]
Abstract
Tumor angiogenesis is mainly regulated by VEGFR-2. In this study, a new series of [1,2,4]triazolo[4,3-a]quinoxaline based-derivatives has been designed and synthesized to develop new anti-proliferative and anti-VEGFR-2 members. Anti-proliferative activities of the synthesized compounds were tested against MCF-7 and HepG2 cell lines. Compound 19a exhibited the highest activity towards both MCF-7 and HepG2 cell lines (IC50 = 8.2 and 5.4 µM, respectively), compared to sorafenib (IC50 = 3.51 and 2.17 µM, respectively). Additionally, all compounds were screened to evaluate their effect as VEGFR-2 inhibitors. Compound 19a (IC50 = 3.4 nM) exhibited good activity compared to sorafenib (IC50 = 3.12 nM). Furthermore, compound 19a disrupted the HepG2 cell cycle by arresting the G2/M phase. Also, marked increase in the percentage apoptotic cells was achieved by compound 19a. The induced apoptotic effect of compound 19a in HepG2 cells was assured by increased pro-apoptotic marker (Bax) expression by 2.33-fold and decreased anti-apoptotic (Bcl-2) expression by 1.88-fold, resulting in an elevation of the Bax/Bcl-2 ratio in HepG2 cells. Comparing to the control cells, compound 19a induced an increase in expression of cleaved caspase-3 and caspase-9 by 2.44- and 2.69-fold, respectively. Finally, the binding modes of the target derivatives were investigated through docking studies against the proposed molecular target (VEGFR-2, PDB ID: 2OH4).
Collapse
Affiliation(s)
- Nawaf A Alsaif
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed S Taghour
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt.
| | - Mohammed M Alanazi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ahmad J Obaidullah
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Wael A Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11541, Saudi Arabia
| | - Abdullah Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11541, Saudi Arabia
| | - Hussam Albassam
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11541, Saudi Arabia
| | - Mohammed A Dahab
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt.
| | - Hazem A Mahdy
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt.
| |
Collapse
|
18
|
Chemical constituents from basidiomycete Basidioradulum radula culture medium and their cytotoxic effect on human prostate cancer DU-145 cells. Bioorg Chem 2021; 114:105064. [PMID: 34134032 DOI: 10.1016/j.bioorg.2021.105064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 05/12/2021] [Accepted: 06/02/2021] [Indexed: 11/21/2022]
Abstract
Eight new naphtho[1,2-c]furan derivatives (1-8) along with six known analogues (9-14) were isolated from culture medium of the basidiomycete Basidioradulum radula. The structures of these compounds were identified using spectroscopic analysis, and their absolute configurations were resolved using X-ray diffraction, ECD, and VCD. Compounds 7 and 14 inhibited the cell viability of human prostate cancer DU-145 cells with IC50 values of 7.54 ± 0.03 μM and 5.04 ± 0.03 μM, respectively. At 8 μM, compounds 7 and 14 increased the percentage of apoptotic cells and upregulated the protein expression related to the apoptosis caspase pathways in DU-145 cells. Furthermore, the hallmarks of cells undergoing apoptosis, such as chromatin condensation, were also observed at this concentration. However, compound 7 and 14 showed no effect on the proliferation of splenocytes isolated from cyclophosphamide-induce immunosuppressed mice.
Collapse
|
19
|
Balakrishnan N, Haribabu J, Dhanabalan AK, Swaminathan S, Sun S, Dibwe DF, Bhuvanesh N, Awale S, Karvembu R. Thiosemicarbazone(s)-anchored water soluble mono- and bimetallic Cu(ii) complexes: enzyme-like activities, biomolecular interactions, anticancer property and real-time live cytotoxicity. Dalton Trans 2021; 49:9411-9424. [PMID: 32589180 DOI: 10.1039/d0dt01309a] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The reactions of CuCl2·2H2O with chromone thiosemicarbazone ligands containing a -H or -CH3 substituent on terminal N yielded monometallic Cu(ii) complexes [Cu(HL1)Cl2] (1) and [Cu(HL2)Cl2] (2), whereas bimetallic Cu(ii) complexes [Cu(μ-Cl)(HL3)]2Cl2 (3), [Cu(μ-Cl)(HL4)]2Cl2 (4) and [Cu(μ-Cl)(L5)]2 (5) were obtained when a -C2H5, -C6H11 or -C6H5 substituent was present, respectively, in the ligands. The complexes were characterized using elemental analyses, UV-Vis, FT-IR, EPR, mass and TGA studies. The structures of neutral monometallic and dicationic bimetallic complexes were confirmed by single crystal X-ray diffraction, and they exhibited a distorted square pyramidal geometry around Cu(ii) ions. The catecholase-mimicking activity of complexes 1-5 was examined spectrophotometrically, and the results revealed that all the complexes except 5 had the ability to oxidize 3,5-di-tert-butylcatechol (3,5-DTBC) to 3,5-di-tert-butylquinone (3,5-DTBQ) under aerobic conditions with moderate turnover numbers. In order to find the possible complex-substrate intermediates, a mass spectrometry study was carried out for complexes 1-4 in the presence of 3,5-DTBC. The phosphatase-like activity of 1-5 was also investigated using 4-nitrophenylphosphate (4-NPP) as a model substrate. All the complexes exhibited excellent phosphatase activity in DMF-H2O medium. The complexes displayed significant biomolecular interactions and antioxidant potential. Complex 3 showed good interaction with apoptotic CASP3 protein, VEGFR2 and PIM-1 kinase receptors as revealed by a molecular docking study. Complexes (3-5) exhibited promising cytotoxicity against HeLa-cervical cancer cells with IC50 values of 2.24 (3), 2.25 (4) and 3.77 (5) μM, respectively, and showed a two-fold higher activity than cisplatin. The active complex 3 showed complete inhibition of colony formation at 10 μM concentration. In addition, the acridine orange (AO)/ethidium bromide (EB) staining and real-time live cell imaging results confirmed that complex 3 induced cell death in HeLa cells.
Collapse
Affiliation(s)
- Nithya Balakrishnan
- Department of Chemistry, National Institute of Technology, Tiruchirappalli 620015, India.
| | - Jebiti Haribabu
- Department of Chemistry, National Institute of Technology, Tiruchirappalli 620015, India. and Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda 278-8510, Japan
| | - Ananda Krishnan Dhanabalan
- Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Guindy Campus, Chennai 600025, India
| | - Srividya Swaminathan
- Department of Chemistry, National Institute of Technology, Tiruchirappalli 620015, India.
| | - Sijia Sun
- Division of Natural Drug Discovery, Department of Translational Research, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Dya Fita Dibwe
- Division of Natural Drug Discovery, Department of Translational Research, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Nattamai Bhuvanesh
- Department of Chemistry, Texas A & M University, College Station, TX 77842, USA
| | - Suresh Awale
- Division of Natural Drug Discovery, Department of Translational Research, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Ramasamy Karvembu
- Department of Chemistry, National Institute of Technology, Tiruchirappalli 620015, India.
| |
Collapse
|
20
|
Apoptosis-Inducing TNF Superfamily Ligands for Cancer Therapy. Cancers (Basel) 2021; 13:cancers13071543. [PMID: 33801589 PMCID: PMC8036978 DOI: 10.3390/cancers13071543] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/21/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer is a complex disease with apoptosis evasion as one of its hallmarks; therefore, apoptosis induction in transformed cells seems a promising approach as a cancer treatment. TNF apoptosis-inducing ligands, which are naturally present in the body and possess tumoricidal activity, are attractive candidates. The most studied proteins are TNF-α, FasL, and TNF-related apoptosis-inducing ligand (TRAIL). Over the years, different recombinant TNF family-derived apoptosis-inducing ligands and agonists have been designed. Their stability, specificity, and half-life have been improved because most of the TNF ligands have the disadvantages of having a short half-life and affinity to more than one receptor. Here, we review the outlook on apoptosis-inducing ligands as cancer treatments in diverse preclinical and clinical stages and summarize strategies of overcoming their natural limitations to improve their effectiveness.
Collapse
|
21
|
Fang L, Wang H, Zhang J, Fang X. Punicalagin induces ROS-mediated apoptotic cell death through inhibiting STAT3 translocation in lung cancer A549 cells. J Biochem Mol Toxicol 2021; 35:1-10. [PMID: 33720461 DOI: 10.1002/jbt.22771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 12/08/2020] [Accepted: 03/02/2021] [Indexed: 12/30/2022]
Abstract
Lung cancer is a noxious disease with substandard overall survival. Despite this, there are several treatment strategies for lung cancer include chemotherapy, radiotherapy, surgery; however, the overall survival remains poor. Punicalagin has been documented as a potential phytomedicine to selectively inhibit the progression and expansion of numerous cancers. In the present study, we evaluated the antiproliferative ability of punicalagin against lung cancer A549 cells by inducing apoptosis by inhibiting STAT-3 activation. Punicalagin induces toxic effects of A549 cells in a dose-associated manner after 24 h treatment. And we also observed that punicalagin (10, 20, and 30 μM) induced reactive oxygen species generation, alters the mitochondrion membrane potential and apoptotic morphological changes in A549 cells. The STAT-3 overexpression regulates apoptosis, proliferation, and angiogenesis. Here, the punicalagin inhibited STAT-3 translocation and thereby induces apoptosis by inhibiting expression Bcl-2 and enhanced expression of Bax, cytochrome-c, caspase-9, and caspase-3 in A549 cells. Hence, we stated that the punicalagin is a possible therapy for non-small cell lung, malignancies. Altogether, the punicalagin is a promising phytomedicine in malignancy treatment and further endeavors are needed to unveil the complete potential.
Collapse
Affiliation(s)
- Le Fang
- Department of Clinical Laboratory, The 521 Hospital of Ordnance Industry, Xi'an, Shaanxi, China.,Department of Blood Transfusion, Institute for Hygiene of Ordnance Industry, Xian, Shaanxi, China
| | - Hong Wang
- Laboratory of Toxicology and Biological Effect, Institute for Hygiene of Ordnance Industry, Xian, Shaanxi, China
| | - Jie Zhang
- Department of Blood Transfusion, Ankang City Central Hospital, Ankang, Shaanxi, China
| | - Xiaolei Fang
- Department of Blood Transfusion, Ankang City Central Hospital, Ankang, Shaanxi, China
| |
Collapse
|
22
|
Pourhajibagher M, Etemad-Moghadam S, Alaeddini M, Bahador A. Modulation of the triggered apoptosis by nano emodin transfersome-mediated sonodynamic therapy on head and neck squamous cell carcinoma cell lines. Photodiagnosis Photodyn Ther 2021; 34:102253. [PMID: 33711532 DOI: 10.1016/j.pdpdt.2021.102253] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/14/2021] [Accepted: 03/05/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Non-invasive sonodynamic therapy (SDT) is a new treatment modality that uses low-intensity ultrasound to activate a non-toxic sensitizing chemical agent for cancer therapy in a site-directed manner. This study aimed to investigate the anti-cancer effects of ultrasound combined with nano emodin transfersome (NET) on head and neck squamous cell carcinoma (HNSCC) cell lines. MATERIALS AND METHODS A transfersome form of nano emodin as a novel sono-responsive nanomaterial was synthesized to enhance the accumulation and penetration of nanoparticles. iIn vitro experiments including hemolytic activity, cell proliferation, intracellular reactive oxygen species (ROS) generation, apoptosis induction, DNA fragmentation, and mRNA expressions of caspase 3 and 9 were conducted to explore the anti-cancer effects of NET-SDT on FaDu and CAL-27 cell lines. RESULTS Characterization tests showed the round and uniform morphology of NET with transfersome structure, resulting in a high drug-loading content and encapsulation efficiency. No significant hemolytic activity was observed (P > 0.05). Cytotoxicity gradually increased with increasing concentrations of NET, so that 10 × 10-4 g/L of NET plus 5 min ultrasound irradiation at a frequency of 1 MHz and ultrasonic intensity of 2 W/cm2 effectively killed 98.2 % and 97.3 % of FaDu and CAL-27 cell lines, respectively (P < 0.05). We found that ROS generation in NET-SDT was dose-dependent and the triggered apoptosis and caspase-3/9 gene expression levels were significantly enhanced as the concentration of NET increased (P < 0.05). No significant difference was found in the rate of apoptosis induction and gene expression between two cell lines. CONCLUSIONS Our data demonstrated that SDT with NET as a sonosensitizer can induce apoptosis and significantly decrease cell viability of HNSCC cell lines, which represents the role of NET-SDT as a potent anti-cancer modality.
Collapse
Affiliation(s)
- Maryam Pourhajibagher
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahroo Etemad-Moghadam
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mojgan Alaeddini
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Bahador
- Oral Microbiology Laboratory, Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Fellowship in Clinical Laboratory Sciences, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
Chen YF, Chang CH, Hsu MW, Chang HM, Chen YC, Jiang YS, Jan JS. Peptide Fibrillar Assemblies Exhibit Membranolytic Effects and Antimetastatic Activity on Lung Cancer Cells. Biomacromolecules 2020; 21:3836-3846. [PMID: 32790281 DOI: 10.1021/acs.biomac.0c00911] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cancer metastasis is a central oncology concern that worsens patient conditions and increases mortality in a short period of time. During metastatic events, mitochondria undergo specific physiological alterations that have emerged as notable therapeutic targets to counter cancer progression. In this study, we use drug-free, cationic peptide fibrillar assemblies (PFAs) formed by poly(L-Lysine)-block-poly(L-Threonine) (Lys-b-Thr) to target mitochondria. These PFAs interact with cellular and mitochondrial membranes via electrostatic interactions, resulting in membranolysis. Charge repulsion and hydrogen-bonding interactions exerted by Lys and Thr segments dictate the packing of the peptides and enable the PFAs to display enhanced membranolytic activity toward cancer cells. Cytochrome c (cyt c), endonuclease G, and apoptosis-inducing factor were released from mitochondria after treatment of lung cancer cells, subsequently inducing caspase-dependent and caspase-independent apoptotic pathways. A metastatic xenograft mouse model was used to show how the PFAs significantly suppressed lung metastasis and inhibited tumor growth, while avoiding significant body weight loss and mortality. Antimetastatic activities of PFAs are also demonstrated by in vitro inhibition of lung cancer cell migration and clonogenesis. Our results imply that the cationic PFAs achieved the intended and targeted mitochondrial damage, providing an efficient antimetastatic therapy.
Collapse
Affiliation(s)
- Yu-Fon Chen
- Department of Chemical Engineering, National Cheng Kung University, No. 1 University Road, Tainan 70101 Taiwan
| | - Chien-Hsiang Chang
- Department of Chemical Engineering, National Cheng Kung University, No. 1 University Road, Tainan 70101 Taiwan
| | - Ming-Wei Hsu
- Department of Chemical Engineering, National Cheng Kung University, No. 1 University Road, Tainan 70101 Taiwan
| | - Ho-Min Chang
- Department of Chemical Engineering, National Cheng Kung University, No. 1 University Road, Tainan 70101 Taiwan
| | - Yi-Cheng Chen
- Department of Chemical Engineering, National Cheng Kung University, No. 1 University Road, Tainan 70101 Taiwan
| | - Yi-Sheng Jiang
- Department of Chemical Engineering, National Cheng Kung University, No. 1 University Road, Tainan 70101 Taiwan
| | - Jeng-Shiung Jan
- Department of Chemical Engineering, National Cheng Kung University, No. 1 University Road, Tainan 70101 Taiwan
| |
Collapse
|
24
|
Shaikh S, Younis M, Rehman FU, Jiang H, Wang X. Specific Oxide Nanoclusters Enhance Intracellular Reactive Oxygen Species for Cancer-Targeted Therapy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:9472-9480. [PMID: 32701296 DOI: 10.1021/acs.langmuir.0c01378] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Bio-nanotechnology based cancer therapeutics exponentially increase every year. A therapeutic strategy to induce intracellular reactive oxygen species (ROS) has received promising success in oncotherapy. In this study, the new strategy has been exploited by the treatment of iridium (Ir) and Fe2+ ions with cancer cells to biosynthesize the biocompatible fluorescent iridium oxide (IrO2) and iron oxide nanoclusters (NCs) under the specific redox heterogeneous microenvironment of these diseased cells and tumors. The hydroxyl radical produced by the presence of Fe2+ and H2O2 in cancer cells apparently increased the ROS level in cancer cells during the process of biosynthesized NCs and, hence, simultaneously instigated apoptosis of relevant cells. Therefore, intracellular ROS-mediated in situ biosynthesis of IrO2 and iron oxide NCs may also act as anticancer agents and provide a promising pathway for targeted cancer therapy.
Collapse
Affiliation(s)
- Sana Shaikh
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, People's Republic of China
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, Jiangsu 210096, People's Republic of China
| | - Muhammad Younis
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, Jiangsu 210096, People's Republic of China
| | - Fawad Ur Rehman
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, People's Republic of China
| | - Hui Jiang
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, People's Republic of China
| | - Xuemei Wang
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, People's Republic of China
| |
Collapse
|
25
|
Dong Z, Hu H, Yu X, Tan L, Ma C, Xi X, Li L, Wang L, Zhou M, Chen T, Du S, Lu Y. Novel Frog Skin-Derived Peptide Dermaseptin-PP for Lung Cancer Treatment: In vitro/vivo Evaluation and Anti-tumor Mechanisms Study. Front Chem 2020; 8:476. [PMID: 32582642 PMCID: PMC7291860 DOI: 10.3389/fchem.2020.00476] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 05/07/2020] [Indexed: 01/10/2023] Open
Abstract
Lung cancer is the major cause of cancer deaths worldwide, and it has the highest incidence and mortality rate of any cancer among men and women in China. The first-line therapy for lung cancer treatment is platinum-based chemotherapy drugs such as cisplatin. However, the application of present chemotherapies is limited by severe side effects, which stimulates the discovery of new drugs with new anti-tumor mechanisms and fewer side effects. Beneficially, many antimicrobial peptides (AMPs) from frog skin have been reported to exhibit potent anti-cancer activities with low toxicity, high selectivity and a low propensity to induce resistance. In this study, we first reported an AMP named Dermaseptin-PP, from a rarely studied frog species, Phyllomedusa palliata. Dermaseptin-PP exhibited selective cytotoxicity on H157, MCF-7, PC-3, and U251 MG cancer cells instead of normal HMEC-1 cells with low hemolytic effect. Furthermore, on subcutaneous H157 tumor model of nude mice, Dermaseptin-PP was found to display potent in vivo anti-tumor activity in a dose-related manner without obvious hepatopulmonary side effects. It is widely accepted that AMPs usually work through a membrane disruptive mode, and the confocal laser microscope observation confirmed that Dermaseptin-PP could destroy H157 cell membranes. Further investigation of mechanisms by flow cytometry assay and immunohistochemical analysis unraveled that Dermaseptin-PP also exerted its anti-tumor activity by inducing H157 cell apoptosis via both endogenous mitochondrial apoptosis pathway and exogenous death receptor apoptosis pathway. Herein, we emphasize that the membrane disrupting and the apoptosis activation effects of Dermaseptin-PP both depend on its concentration. Overall, a novel frog skin-derived AMP, named Dermaseptin-PP, was identified for the first time. It possesses strong antimicrobial activity and effective anti-tumor activity by distinct mechanisms. This study revealed the possibility of Dermaseptin-PP for lung cancer treatment and provided a new perspective for designing novel AMP-based anti-tumor candidates with low risk of cytotoxicity.
Collapse
Affiliation(s)
- Ziyi Dong
- Laboratory of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Haiyan Hu
- Laboratory of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xianglong Yu
- Laboratory of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Li Tan
- Livzon Pharmaceutical Group Inc., Zhuhai, China
| | - Chengbang Ma
- Natural Drug Discovery Group, School of Pharmacy, Queen's University, Belfast, United Kingdom
| | - Xinping Xi
- Natural Drug Discovery Group, School of Pharmacy, Queen's University, Belfast, United Kingdom
| | - Lei Li
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Lei Wang
- Natural Drug Discovery Group, School of Pharmacy, Queen's University, Belfast, United Kingdom
| | - Mei Zhou
- Natural Drug Discovery Group, School of Pharmacy, Queen's University, Belfast, United Kingdom
| | - Tianbao Chen
- Natural Drug Discovery Group, School of Pharmacy, Queen's University, Belfast, United Kingdom
| | - Shouying Du
- Laboratory of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Lu
- Laboratory of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
26
|
Gano L, Pinheiro T, Matos AP, Tortosa F, Jorge TF, Gonçalves MS, Martins M, Morais TS, Valente A, Tomaz AI, Garcia MH, Marques F. Antitumour and Toxicity Evaluation of a Ru(II)-Cyclopentadienyl Complex in a Prostate Cancer Model by Imaging Tools. Anticancer Agents Med Chem 2020; 19:1262-1275. [PMID: 30887931 DOI: 10.2174/1871520619666190318152726] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/19/2018] [Accepted: 03/06/2019] [Indexed: 01/24/2023]
Abstract
BACKGROUND Ruthenium complexes have been extensively investigated for their prospective value as alternatives to cisplatin. Recently, we reported the in vitro anticancer properties of a family of organometallic ruthenium( II)-cyclopentadienyl complexes and have explored their mechanism of action. OBJECTIVE The purpose of this study was to evaluate the in vivo antitumour efficacy and toxicity of one of these Ru(II) compounds, [RuCp(mTPPMSNa)(2,2'-bipy)][CF3SO2] (TM85) which displayed an interesting spectrum of activity against several cancer cells. METHODS Studies to assess the antitumour activity and toxicity were performed in a metastatic prostate (PC3) mice model using ICP-MS, nuclear microscopy, elemental analysis and Transmission Electron Microscopy (TEM). RESULTS TM85 showed low systemic toxicity but no significant tumour reduction, when administered at tolerated dose (20mg/kg) over 10 days. Ru was mainly retained in the liver and less in kidneys, with low accumulation in tumour. Increased bilirubin levels, anomalous Ca and Fe concentrations in liver and mitochondria alterations were indicative of liver injury. The hepatotoxicity observed was less severe than that of cisplatin and no nephrotoxicity was found. CONCLUSION Under the experimental conditions of this study, TM85 is less toxic than cisplatin, induces similar tumour reduction and avoids the formation of metastatic foci. No renal toxicity was observed by the analysis of creatinine levels and the effective renal plasma flow by 99mTc-MAG3 clearance. Hence, it can be considered a valuable compound for further studies in the field of Ru-based anticancer drugs.
Collapse
Affiliation(s)
- Lurdes Gano
- Centro de Ciencias e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, km 139.7, 2695-066 Bobadela LRS, Lisboa, Portugal
| | - Teresa Pinheiro
- Departamento de Engenharia e Ciencias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, km 139.7, 2695-066 Bobadela LRS, Lisboa, Portugal
| | - António P Matos
- Centro de Investigacao Interdisciplinar Egas Moniz, Campus Universitario, Quinta da Granja, Monte de Caparica, 2829-511 Caparica, Portugal
| | - Francisco Tortosa
- Instituto de Anatomia Patologica, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal.,Departamento de Medicina, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Tiago F Jorge
- Centro de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal.,Laboratório de Metabolómica de Plantas, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Av. da República, 2780-157 Oeiras, Lisboa, Portugal
| | - Maria S Gonçalves
- Faculdade de Medicina Veterinaria, Universidade de Lisboa, Av. da Universidade Tecnica, Polo Universitario da Ajuda 1300-477 Lisboa, Portugal
| | - Marta Martins
- Instituto de Medicina Molecular-Joao Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Tânia S Morais
- Centro de Quimica Estrutural, Faculdade de Ciencias, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Andreia Valente
- Centro de Quimica Estrutural, Faculdade de Ciencias, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Ana I Tomaz
- Centro de Quimica Estrutural, Faculdade de Ciencias, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Maria H Garcia
- Centro de Quimica Estrutural, Faculdade de Ciencias, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Fernanda Marques
- Centro de Ciencias e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, km 139.7, 2695-066 Bobadela LRS, Lisboa, Portugal
| |
Collapse
|
27
|
Iguratimod encapsulated PLGA-NPs improves therapeutic outcome in glioma, glioma stem-like cells and temozolomide resistant glioma cells. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 22:102101. [PMID: 31654739 DOI: 10.1016/j.nano.2019.102101] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/17/2019] [Accepted: 09/25/2019] [Indexed: 01/27/2023]
Abstract
Glioma is the most common neoplasm of the central nervous system, with the highest mortality rate. The present study was designed to examine the therapeutic effect of Iguratimod (IGU) encapsulated-poly (lactic-co-glycolic acid) PLGA nanoparticles (IGU-PLGA-NPs), which showed inhibition of glioma cells proliferation both in vitro and in vivo. IGU encapsulated in PLGA nanoparticles with an average size of 100-200 nm was prepared using modified double-emulsion (W1/O/W2) method. Cell Counting Kit-8 (CCK-8) analysis of Glioma cancer cells and glioma stem-like cells (GSCs) demonstrated significant inhibition of their growth treated with IGU-PLGA-NPs. IGU-PLGA-NPs inhibit migration in glioma cells as well as tumor sphere formation in GSCs. Treatment with IGU-PLGA-NPs showed a significant decrease in tumor growth through the apoptotic pathway in mice model without any visible organ toxicity and it can successfully cross the blood brain barrier (BBB). Most Importantly, IGU-PLGA-NPs significantly depleted growth of U251 Temozolomide-resistant (U251TMZ-R) cells.
Collapse
|
28
|
Adamska A, Stefanowicz-Hajduk J, Ochocka JR. Alpha-Hederin, the Active Saponin of Nigella sativa, as an Anticancer Agent Inducing Apoptosis in the SKOV-3 Cell Line. Molecules 2019; 24:molecules24162958. [PMID: 31443189 PMCID: PMC6719954 DOI: 10.3390/molecules24162958] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/10/2019] [Accepted: 08/13/2019] [Indexed: 12/31/2022] Open
Abstract
Alpha-hederin (α-HN), a pentacyclic triterpene saponin, has recently been identified as one of the active compounds of Nigella sativa, as a potential anticancer agent. However, no extensive studies on α-HN have been done as yet, as it was in the case of thymoquinone—the main ingredient of the N.sativa essential oil. To our knowledge, there are also no data available on how α-HN acts on the human cancer ovarian cell line SKOV-3. In this study we attempt to present the cytotoxic influence of α-HN on the SKOV-3 cell line by means of two methods: Real-Time xCELLigence and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The obtained IC50 values are 2.62 ± 0.04 μg/mL and 2.48 ± 0.32 μg/mL, respectively. An induction of apoptosis in SKOV-3 cells was confirmed by staining cellular nuclei with Hoechst 33342 dye and by flow cytometry analysis by binding annexin V to the cell membranes. We found that α-HN induces apoptosis in a dose-dependent manner. In the first stages of apoptosis, the mitochondrial membrane potential was found to decrease. Also, inactivation of anti-apoptotic protein Bcl-2 was observed, as well as the caspase-9 and then caspase-3/7 activation. In addition, the treatment of SKOV-3 cells with α-HN induced the cell cycle arrest of cancer cells in G0/G1 phase. The results of our investigations indicate that α-HN induces apoptosis in the SKOV-3 cell line and that the intrinsic mitochondrial pathway is involved in the programmed cancer cell death.
Collapse
Affiliation(s)
- Anna Adamska
- Department of Biology and Pharmaceutical Botany, Medical University of Gdańsk, Al. Hallera 107, 80-416 Gdańsk, Poland.
| | - Justyna Stefanowicz-Hajduk
- Department of Biology and Pharmaceutical Botany, Medical University of Gdańsk, Al. Hallera 107, 80-416 Gdańsk, Poland
| | - J Renata Ochocka
- Department of Biology and Pharmaceutical Botany, Medical University of Gdańsk, Al. Hallera 107, 80-416 Gdańsk, Poland.
| |
Collapse
|
29
|
Akhtar S, Achkar IW, Siveen KS, Kuttikrishnan S, Prabhu KS, Khan AQ, Ahmed EI, Sahir F, Jerobin J, Raza A, Merhi M, Elsabah HM, Taha R, Omri HE, Zayed H, Dermime S, Steinhoff M, Uddin S. Sanguinarine Induces Apoptosis Pathway in Multiple Myeloma Cell Lines via Inhibition of the JaK2/STAT3 Signaling. Front Oncol 2019; 9:285. [PMID: 31058086 PMCID: PMC6478801 DOI: 10.3389/fonc.2019.00285] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 03/29/2019] [Indexed: 12/15/2022] Open
Abstract
Sanguinarine (SNG), a benzophenanthridine alkaloid, has displayed various anticancer abilities in several vivo and in vitro studies. However, the anticancer potential of SNG is yet to be established in multiple myeloma (MM), a mostly incurable malignancy of plasma cells. In this study, we aimed to investigate the potential anti-proliferative and pro-apoptotic activities of SNG in a panel of MM cell lines (U266, IM9, MM1S, and RPMI-8226). SNG treatment of MM cells resulted in a dose-dependent decrease in cell viability through mitochondrial membrane potential loss and activation of caspase 3, 9, and cleavage of PARP. Pre-treatment of MM cells with a universal caspase inhibitor, Z-VAD-FMK, prevented SNG mediated loss of cell viability, apoptosis, and caspase activation, confirming that SNG-mediated apoptosis is caspase-dependent. The SNG-mediated apoptosis appears to be resulted from suppression of the constitutively active STAT3 with a concomitant increase in expression of protein tyrosine phosphatase (SHP-1). SNG treatment of MM cells leads to down-regulation of the anti-apoptotic proteins including cyclin D, Bcl-2, Bclxl, and XIAP. In addition, it also upregulates pro-apoptotic protein, Bax. SNG mediated cellular DNA damage in MM cell lines by induction of oxidative stress through the generation of reactive oxygen species and depletion of glutathione. Finally, the subtoxic concentration of SNG enhanced the cytotoxic effects of anticancer drugs bortezomib (BTZ) by suppressing the viability of MM cells via induction of caspase-mediated apoptosis. Altogether our findings demonstrate that SNG induces mitochondrial and caspase-dependent apoptosis, generates oxidative stress, and suppresses MM cell lines proliferation. In addition, co-treatment of MM cell lines with sub-toxic doses of SNG and BTZ potentiated the cytotoxic activity. These results would suggest that SNG could be developed into therapeutic agent either alone or in combination with other anticancer drugs in MM.
Collapse
Affiliation(s)
- Sabah Akhtar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Iman W. Achkar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Kodappully S. Siveen
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Shilpa Kuttikrishnan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Kirti S. Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Abdul Q. Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Eiman I. Ahmed
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Fairooz Sahir
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Jayakumar Jerobin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Afsheen Raza
- Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Maysaloun Merhi
- Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Hesham M. Elsabah
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Ruba Taha
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Halima El Omri
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Hatem Zayed
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar
| | - Said Dermime
- Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Martin Steinhoff
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Department of Dermatology Venereology, Hamad Medical Corporation, Doha, Qatar
- Weill Cornell-Medicine, Doha, Qatar
- Weill Cornell-Medicine, Cornell University, New York, NY, United States
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
30
|
Salah Ayoup M, Wahby Y, Abdel-Hamid H, Ramadan ES, Teleb M, Abu-Serie MM, Noby A. Design, synthesis and biological evaluation of novel α-acyloxy carboxamides via Passerini reaction as caspase 3/7 activators. Eur J Med Chem 2019; 168:340-356. [DOI: 10.1016/j.ejmech.2019.02.051] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/16/2019] [Accepted: 02/17/2019] [Indexed: 12/17/2022]
|
31
|
Kuttikrishnan S, Siveen KS, Prabhu KS, Khan AQ, Akhtar S, Mateo JM, Merhi M, Taha R, Omri HE, Mraiche F, Dermime S, Uddin S. Sanguinarine suppresses growth and induces apoptosis in childhood acute lymphoblastic leukemia. Leuk Lymphoma 2019; 60:782-794. [PMID: 30187808 DOI: 10.1080/10428194.2018.1494270] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/19/2018] [Accepted: 06/24/2018] [Indexed: 12/17/2022]
Abstract
Sanguinarine (Sang), a plant-derived compound isolated from the roots of Sanguinaria canadensis was evaluated for its potential pro-apoptotic effects in precursor B acute lymphoblastic leukemia (Pre-ALL) cell lines. Treatment of 697, REH, RS4;11, and SupB15 cell lines with Sang exhibited significant inhibition of cell viability via induction of apoptotic cell death. Sang-mediated apoptosis was found to be associated with the increased expression of proapoptotic bax with concomitant decrease of Bcl-2 expression leading to depolarization of mitochondria membrane resulting in loss of mitochondrial membrane potential (MMP). The reduced MMP caused the leakage in mitochondrial membrane and release of cytochrome c into the cytosol. The cytochrome c then mediates the activation of caspase-cascade and subsequently PARP cleavage. Furthermore, pretreatment with z-VAD-FMK, a pan-caspase inhibitor, abrogated Sang-induced inhibition of cell viability, induction of apoptosis. Sang treatment also reduced the phosphorylation of AKT and suppressed the expression of a number of anti-apoptotic genes such as cIAP1, cIAP2, and XIAP. Sang mediates its anti-cancer activity by generation of reactive oxygen species (ROS) due to depletion of glutathione level in leukemic cell lines. Pretreatment of these cells with N-acetyl cysteine (NAC) prevented Sang-induced depletion of glutathione level and mitochondrial-caspase-induced apoptosis. Finally, Sang treatment of Pre-ALL cell suppressed colony formation ability of these cells suggesting Sang has an anti-leukemic potential. Altogether, our data suggest that Sang is an efficient inducer of intrinsic apoptotic cell death via generation of ROS and exhibition of anti-leukemic effect in Pre-ALL cells raises the possibility to develop Sang as a therapeutic modality for the treatment and management of Pre-ALL.
Collapse
Affiliation(s)
- Shilpa Kuttikrishnan
- a Translational Research Institute , Academic Health System, Hamad Medical Corporation , Doha , Qatar
| | - Kodappully S Siveen
- a Translational Research Institute , Academic Health System, Hamad Medical Corporation , Doha , Qatar
| | - Kirti S Prabhu
- a Translational Research Institute , Academic Health System, Hamad Medical Corporation , Doha , Qatar
| | - Abdul Quaiyoom Khan
- a Translational Research Institute , Academic Health System, Hamad Medical Corporation , Doha , Qatar
| | - Sabah Akhtar
- a Translational Research Institute , Academic Health System, Hamad Medical Corporation , Doha , Qatar
| | - Jericha M Mateo
- a Translational Research Institute , Academic Health System, Hamad Medical Corporation , Doha , Qatar
| | - Maysaloun Merhi
- b National Centre for Cancer Care and Research , Hamad Medical Corporation , Doha , Qatar
| | - Ruba Taha
- b National Centre for Cancer Care and Research , Hamad Medical Corporation , Doha , Qatar
| | - Halima El Omri
- b National Centre for Cancer Care and Research , Hamad Medical Corporation , Doha , Qatar
| | | | - Said Dermime
- b National Centre for Cancer Care and Research , Hamad Medical Corporation , Doha , Qatar
| | - Shahab Uddin
- a Translational Research Institute , Academic Health System, Hamad Medical Corporation , Doha , Qatar
| |
Collapse
|
32
|
Fromm G, de Silva S, Johannes K, Patel A, Hornblower JC, Schreiber TH. Agonist redirected checkpoint, PD1-Fc-OX40L, for cancer immunotherapy. J Immunother Cancer 2018; 6:149. [PMID: 30563566 PMCID: PMC6299665 DOI: 10.1186/s40425-018-0454-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 11/16/2018] [Indexed: 12/19/2022] Open
Abstract
Simultaneous blockade of immune checkpoint molecules and co-stimulation of the TNF receptor superfamily (TNFRSF) is predicted to improve overall survival in human cancer. TNFRSF co-stimulation depends upon coordinated antigen recognition through the T cell receptor followed by homotrimerization of the TNFRSF, and is most effective when these functions occur simultaneously. To address this mechanism, we developed a two-sided human fusion protein incorporating the extracellular domains (ECD) of PD-1 and OX40L, adjoined by a central Fc domain, termed PD1-Fc-OX40L. The PD-1 end of the fusion protein binds PD-L1 and PD-L2 with affinities of 2.08 and 1.76 nM, respectively, and the OX40L end binds OX40 with an affinity of 246 pM. High binding affinity on both sides of the construct translated to potent stimulation of OX40 signaling and PD1:PD-L1/L2 blockade, in multiple in vitro assays, including improved potency as compared to pembrolizumab, nivolumab, tavolixizumab and combinations of those antibodies. Furthermore, when activated human T cells were co-cultured with PD-L1 positive human tumor cells, PD1-Fc-OX40L was observed to concentrate to the immune synapse, which enhanced proliferation of T cells and production of IL-2, IFNγ and TNFα, and led to efficient killing of tumor cells. The therapeutic activity of PD1-Fc-OX40L in established murine tumors was significantly superior to either PD1 blocking, OX40 agonist, or combination antibody therapy; and required CD4+ T cells for maximum response. Importantly, all agonist functions of PD1-Fc-OX40L are independent of Fc receptor cross-linking. Collectively, these data demonstrate a highly potent fusion protein that is part of a platform, capable of providing checkpoint blockade and TNFRSF costimulation in a single molecule, which uniquely localizes TNFRSF costimulation to checkpoint ligand positive tumor cells.
Collapse
Affiliation(s)
- George Fromm
- Shattuck Labs, Inc, 21 Parmer Way, Suite 200, Durham, NC, 27703, USA
| | - Suresh de Silva
- Shattuck Labs, Inc, 21 Parmer Way, Suite 200, Durham, NC, 27703, USA
| | - Kellsey Johannes
- Shattuck Labs, Inc, 21 Parmer Way, Suite 200, Durham, NC, 27703, USA
| | - Arpita Patel
- Shattuck Labs, Inc, 21 Parmer Way, Suite 200, Durham, NC, 27703, USA
| | | | | |
Collapse
|
33
|
Prabhu KS, Siveen KS, Kuttikrishnan S, Iskandarani AN, Khan AQ, Merhi M, Omri HE, Dermime S, El-Elimat T, Oberlies NH, Alali FQ, Uddin S. Greensporone C, a Freshwater Fungal Secondary Metabolite Induces Mitochondrial-Mediated Apoptotic Cell Death in Leukemic Cell Lines. Front Pharmacol 2018; 9:720. [PMID: 30061828 PMCID: PMC6054921 DOI: 10.3389/fphar.2018.00720] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 06/13/2018] [Indexed: 12/17/2022] Open
Abstract
Therapeutic agents used in the treatment of cancer are known to develop resistance against cancer cells. Hence, there is a continuing need to investigate novel agents for the treatment and management of cancer. Antitumor activity of greensporone C (GC), a new resorcylic acid lactone isolated from an organic extract of a culture of a Halenospora sp. freshwater fungus, was subjected for screening against a panel of leukemic cell lines (K562, U937, and AR320). In all the three cell lines, cell proliferation was inhibited in dose-dependent fashion. GC further arrested the cells in SubG0 phase in dose-dependent manner. Annexin V/PI dual staining data confirmed apoptotic death of treated K562 and U937 leukemic cells. Treatment with GC suppressed constitutively phosphorylated AKT and downregulated expression of inhibitor of apoptotic proteins XIAP, cIAP-1, and cIAP-2. In summation to this, GC-treated leukemic cells upregulated protein expression of pro-apoptotic proteins, Bax with concomitant decrease in expression of anti-apoptotic proteins including Bcl-2 and Bcl-xL. Upregulation of Bax was associated with cytochrome c release which was confirmed from the collapse of mitochondrial membrane. Released cytochrome c further activated caspase cascade which in turn initiated apoptosis process. Anticancer activity of this isolated fungal compound GC was potentiated via stimulating production of reactive oxygen species (ROS) along with depletion of reduced glutathione (GSH) levels in K562 and U937 leukemic cells. Pretreatment of these cells with N-acetyl cysteine prevented GC-induced depletion of reduced GSH level and mitochondrial-caspase-induced apoptosis. Altogether, our data show that GC modulates the apoptotic response of human leukemic cells and raises the possibility of its use as a novel therapeutic strategy for hematological malignancies.
Collapse
Affiliation(s)
- Kirti S. Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | | | - Shilpa Kuttikrishnan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Ahmad N. Iskandarani
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Abdul Q. Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Maysaloun Merhi
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Halima E. Omri
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Said Dermime
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Tamam El-Elimat
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Nicholas H. Oberlies
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC, United States
| | | | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
34
|
Banete A, Seaver K, Bakshi D, Gee K, Basta S. On taking the STING out of immune activation. J Leukoc Biol 2018; 103:1189-1195. [PMID: 29431896 DOI: 10.1002/jlb.2mir0917-383r] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 01/17/2018] [Indexed: 12/18/2022] Open
Abstract
Nearly a decade ago, an endoplasmic reticulum (ER) adaptor protein called stimulator of interferon genes (STING) was found to be critical in the induction of type I IFN production in response to DNA virus infection. STING functions by sensing cytoplasmic DNA and activates key transcription factors, including IFN regulatory factor (IRF)-3 and IRF7, to initiate type I IFN expression. Type I IFNs are vital in immunity against viral infections and can influence cancer cell proliferation, migration, and apoptosis. Several studies have shown that STING activation results in potent antitumor activity by generating strong tumor-specific cytotoxic T-cell responses. Moreover, compared with wild-type, STING-knockout mice show greater susceptibility to viral infections. In this review, we discuss the importance of STING signaling during the induction of immune responses, especially those associated with type I IFN in viral infections and tumor immunity. Furthermore, we highlight recent data that unravel how the STING signaling pathway can be negatively regulated.
Collapse
Affiliation(s)
- Andra Banete
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Kyle Seaver
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Devyani Bakshi
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Katrina Gee
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Sameh Basta
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
35
|
Bonavida B, Chouaib S. Resistance to anticancer immunity in cancer patients: potential strategies to reverse resistance. Ann Oncol 2017; 28:457-467. [PMID: 27864216 DOI: 10.1093/annonc/mdw615] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In the 1990s, the application of immunotherapy approaches to target cancer cells resulted in significant clinical responses in patients with advanced malignancies who were refractory to conventional therapies. While early immunotherapeutics were focused on T cell-mediated cytotoxic activity, subsequent efforts were centered on targeted antibody-mediated anticancer therapy. The initial success with antibody therapy encouraged further studies and, consequently, there are now more than 25 FDA-approved antibodies directed against a range of targets. Although both T cell and antibody therapies continue to result in significant clinical responses with minimal toxicity, a significant subset of patients does not respond to immunotherapy and another subset develops resistance following an initial response. This review is focused on describing examples showing that cancer resistance to immunotherapies indeed occurs. In addition, it reviews the mechanisms being used to overcome the resistance to immunotherapies by targeting the tumor cell directly and/or the tumor microenvironment.
Collapse
Affiliation(s)
- B Bonavida
- Department of Microbiology, Immunology and Molecular Genetics, Jonsson Comprehensive Cancer Center and David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, USA
| | - S Chouaib
- Institut de Cancérologie Gustave Roussy, Inserm U1186, Immunologie Intégrative et Oncogénétique, Institut Gustave Roussy, Université Paris-Sud, Université Paris-Saclay Villejuif, France
| |
Collapse
|
36
|
Fatemi Naeini F, Abtahi-Naeini B, Najafian J, Saffaei A, Pourazizi M. Correlation between mycosis fungoides and pregnancy. Saudi Med J 2017; 37:968-72. [PMID: 27570852 PMCID: PMC5039616 DOI: 10.15537/smj.2016.9.15838] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Objectives: To evaluate the effect of pregnancy on the natural course of Mycosis fungoides (MF) and compare the obtained results with previous reports. Methods: The medical records of 140 patients with cutaneous T-cell lymphoma (CTCL) treated at the University Hospital of Isfahan (the academic referral center for CTCL) Isfahan, Iran. Between 2000 and 2013 were retrospectively reviewed to retrieve all cases of pregnancy during the course of MF disease. Results: A total of 8 pregnancies were recorded. The median age of patients at the time of diagnosis was 26.7 (range 21-30 years) and pregnancy 29.4 (range 27-31 years). Most of patients had early-stage MF (Ia and Ib). All patients experienced aggravation of disease during pregnancy or immediately postpartum. Mycosis fungoides did not cause any complications during pregnancy. Conclusion: Pregnancy appears to have a negative impact on the course of MF, probably due to immune system deteriorations during the pregnancy. Further studies are needed to clarify the interplay between pregnancy and MF.
Collapse
Affiliation(s)
- Farahnaz Fatemi Naeini
- Department of Dermatology, Skin Diseases and Leishmaniasis Research Center, Isfahan University of Medical sciences, Isfahan, Iran. E-mail.
| | | | | | | | | |
Collapse
|
37
|
Lee C, Chun W, Zhao R, Kim YD, Nam MM, Jung DH, Cho IJ, Jegal KH, Lee TH, Kim YW, Park SM, Ju SA, Lee CW, Kim SC, An WG. Anticancer effects of an extract from the scallop Patinopecten yessoensis on MCF-7 human breast carcinoma cells. Oncol Lett 2017; 14:2207-2217. [PMID: 28789443 PMCID: PMC5530092 DOI: 10.3892/ol.2017.6424] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 03/03/2017] [Indexed: 01/15/2023] Open
Abstract
Patinopecten yessoensis, is a species of scallop and a marine bivalve mollusk. In traditional East Asian medicine, scallop meat is used as a drug for the treatment of diabetes, pollakisuria, and indigestion. The present study was conducted in order to examine the potential anticancer effects of scallop flesh extract (SE) on MCF-7 human breast cancer cells. An MTT assay was used to evaluate cell viability and flow cytometry was used for the assessment of cell cycle distribution and apoptosis. The alteration in protein expression level was determined by western blot analysis, and the amounts of docosahexaenoic acid and eicosapentaenoic acid in the SE were measured by gas chromatography. SE inhibited the growth of MCF-7 human breast cancer cells in a dose-dependent manner by inducing G0/G1 phase arrest. The cell cycle arrest was associated with the upregulation of p53 and p21, and downregulation of G1 phase-associated cyclin D1/cyclin-dependent kinase (Cdk) 4 and cyclin E1/Cdk 2. In addition, SE-mediated cell cycle arrest was associated with the promotion of apoptosis, as indicated by the expression of apoptosis-associated proteins and changes in nuclear morphology. SE appeared to induce the mitochondrial apoptotic cascade, as indicated by a decreased expression of Bcl-2, activation of Bcl-2 associated X protein, release of cytochrome c, decrease in procaspase-3, and an increase in cleaved-poly (ADP-ribose) polymerase (PARP). Furthermore, the expression levels of Fas-associated via death domain and cleaved caspase-8 were increased in a SE dose-dependent manner. Taken together, these results suggest that the intrinsic and extrinsic pathways of apoptosis are associated with the anticancer effects of SE on MCF-7 cells. Thus, SE may be a suitable candidate for the treatment and prevention of human breast cancer.
Collapse
Affiliation(s)
- Chu Lee
- Aquaculture Industry Division, NFRDI, Gangneung 210-809, Republic of Korea
| | - Wonjoo Chun
- Institute of Marine Biotechnology, Pusan National University, Busan 609-735, Republic of Korea
| | - Rongjie Zhao
- School of Mental Health, Qiqihar Medical University, Qiqihar, Heilongjiang 161042, P.R. China
| | - Young Dae Kim
- Aquaculture Industry Division, NFRDI, Gangneung 210-809, Republic of Korea
| | - Myung Mo Nam
- Aquaculture Industry Division, NFRDI, Gangneung 210-809, Republic of Korea
| | - Dae Hwa Jung
- HaniBio Co., Ltd., Gyeongsan 712-260, Republic of Korea
| | - Il Je Cho
- MRC-GHF, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Kyung Hwan Jegal
- MRC-GHF, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Tae Hoon Lee
- Department of Biological Sciences, College of Biomedical Sciences and Engineering, Inje University, Gimhae 621-749, Republic of Korea
| | - Young Woo Kim
- MRC-GHF, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Sang Mi Park
- MRC-GHF, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Seong A Ju
- School of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Chul Won Lee
- Institute of Marine Biotechnology, Pusan National University, Busan 609-735, Republic of Korea.,MRC-GHF, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Sang Chan Kim
- MRC-GHF, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Won G An
- Institute of Marine Biotechnology, Pusan National University, Busan 609-735, Republic of Korea.,Division of Pharmacology, School of Korean Medicine, Pusan National University, Yangsan 626-870, Republic of Korea
| |
Collapse
|
38
|
Park C, Hong SH, Choi YH. Induction of apoptosis by Dae-Hwang-Mok-Dan-Tang in HCT-116 colon cancer cells through activation of caspases and inactivation of the phosphatidylinositol 3-kinase/Akt signaling. Integr Med Res 2017; 6:179-189. [PMID: 28664141 PMCID: PMC5478258 DOI: 10.1016/j.imr.2017.03.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/21/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Dae-Hwang-Mok-Dan-Tang (DHMDT), a traditional Korean medicine, contains five species of medicinal plants and has been used to treat patients with digestive tract cancer for hundreds of years; however, its anticancer mechanism is poorly understood. In the present study, we investigated the proapoptotic effects of DHMDT in human colon cancer HCT-116 cells. METHODS Cytotoxicity was evaluated using the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide assay. Apoptosis was detected using 4,6-diamidino-2-phenyllindile staining, agarose gel electrophoresis, and flow cytometry. The protein levels were determined using Western blot analysis. Caspase activity was measured using a colorimetric assay. RESULTS Treatment with DHMDT resulted in a growth inhibition coupled with apoptosis induction, which was associated with the downregulation of members of IAP (inhibitor of apoptosis protein) family, including XIAP and survivin, and the activation of caspase-9 and -3 accompanied by proteolytic degradation of poly(ADP-ribose)-polymerase and phospholipase C-γ1. DHMDT treatment also showed a correlation with the translocation of proapoptotic Bax to mitochondria, the loss of mitochondrial membrane permeabilization, and the cytochrome c release from the mitochondria to the cytosol. Moreover, DHMDT increased the levels of death receptor-associated ligands and enhanced activation of caspase-8 and cleavage of its substrate, Bid. However, the pan-caspase inhibitor could reverse DHMDT-induced apoptosis. In addition, DHMDT suppressed the phosphoinositide 3-kinase (PI3K)/Akt pathway, and treatment with a potent inhibitor of PI3K further increased the apoptotic activity of DHMDT. CONCLUSION Our data showed that DHMDT induces HCT-116 cell apoptosis by activating intrinsic and extrinsic apoptosis pathways and by suppressing the PI3K/Akt signal pathway; however, further studies are needed to identify the active compounds.
Collapse
Affiliation(s)
- Cheol Park
- Department of Molecular Biology, College of Natural Sciences, Dongeui University, Busan, Korea
| | - Su Hyun Hong
- Department of Biochemistry, Dongeui University College of Korean Medicine, Busan, Korea
| | - Yung Hyun Choi
- Department of Biochemistry, Dongeui University College of Korean Medicine, Busan, Korea.,Anti-Aging Research Center & Blue-Bio Industry Regional Innovation Center, Dongeui University, Busan, Korea
| |
Collapse
|
39
|
Park C, Han MH, Park SH, Hong SH, Kim GY, Moon SK, Kim WJ, Choi YH. Induction of apoptosis by Moutan Cortex Radicis in human gastric cancer cells through the activation of caspases and the AMPK signaling pathway. REVISTA BRASILEIRA DE FARMACOGNOSIA-BRAZILIAN JOURNAL OF PHARMACOGNOSY 2017. [DOI: 10.1016/j.bjp.2016.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
40
|
Yoon JJ, Jeong JW, Choi EO, Kim MJ, Hwang-Bo H, Kim HJ, Hong SH, Park C, Lee DH, Choi YH. Protective effects of Scutellaria baicalensis Georgi against hydrogen peroxide-induced DNA damage and apoptosis in HaCaT human skin keratinocytes. EXCLI JOURNAL 2017; 16:426-438. [PMID: 28694748 PMCID: PMC5491928 DOI: 10.17179/excli2016-817] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 03/13/2017] [Indexed: 12/18/2022]
Abstract
Oxidative stress due to excessive accumulation of reactive oxygen species (ROS) is one of the risk factors for the development of several chronic diseases. In this study, we investigated the protective effects of Scutellaria baicalensis rhizome ethanol extract (SBRE) against oxidative stress-induced cellular damage and elucidated the underlying mechanisms in the HaCaT human skin keratinocyte cell line. Our results revealed that treatment with SBRE prior to hydrogen peroxide (H2O2) exposure significantly increased viability of HaCaT cells. SBRE also effectively attenuated H2O2-induced comet tail formation and inhibited the H2O2-induced phosphorylation levels of the histone γH2AX, as well as the number of apoptotic bodies and Annexin V-positive cells. In addition, SBRE exhibited scavenging activity against intracellular ROS generation and restored the mitochondrial membrane potential loss by H2O2. Moreover, H2O2 enhanced the cleavage of caspase-3 and degradation of poly (ADP-ribose)-polymerase, a typical substrate protein of activated caspase-3, as well as DNA fragmentation; however, these events were almost totally reversed by pretreatment with SBRE. Furthermore, SBRE increased the levels of heme oxygenase-1 (HO-1), which is a potent antioxidant enzyme, associated with the induction of nuclear factor-erythroid 2-related factor 2 (Nrf2). According to our data, SBRE is able to protect HaCaT cells from H2O2-induced DNA damage and apoptosis through blocking cellular damage related to oxidative stress through a mechanism that would affect ROS elimination and activating the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Jung Jeh Yoon
- Anti-Aging Research Center and Department of Biochemistry, Dongeui University College of Korean Medicine, 176 Yangjeong-ro, Busanjin-gu, Busan 47227, Republic of Korea
| | - Jin-Woo Jeong
- Anti-Aging Research Center and Department of Biochemistry, Dongeui University College of Korean Medicine, 176 Yangjeong-ro, Busanjin-gu, Busan 47227, Republic of Korea
| | - Eun Ok Choi
- Anti-Aging Research Center and Department of Biochemistry, Dongeui University College of Korean Medicine, 176 Yangjeong-ro, Busanjin-gu, Busan 47227, Republic of Korea
| | - Min Ju Kim
- Lioele Cosmetic Co., LTD., 2068-1 Jungangdae-ro, Geumjeong-gu, Busan 46214, Republic of Korea
| | - Hyun Hwang-Bo
- Anti-Aging Research Center and Department of Biochemistry, Dongeui University College of Korean Medicine, 176 Yangjeong-ro, Busanjin-gu, Busan 47227, Republic of Korea
| | - Hong Jae Kim
- Anti-Aging Research Center and Department of Biochemistry, Dongeui University College of Korean Medicine, 176 Yangjeong-ro, Busanjin-gu, Busan 47227, Republic of Korea
| | - Su Hyun Hong
- Anti-Aging Research Center and Department of Biochemistry, Dongeui University College of Korean Medicine, 176 Yangjeong-ro, Busanjin-gu, Busan 47227, Republic of Korea
| | - Cheol Park
- Department of Molecular Biology, College of Natural Sciences, Dongeui University, 176 Eomgwangno Busanjin-gu, Busan 47340, Republic of Korea
| | - Dong Hee Lee
- Genomine Inc., Venture Bldg 306, Pohang TechnoPark, 394 Jigokor Pohang, 37668, Republic of Korea
| | - Yung Hyun Choi
- Anti-Aging Research Center and Department of Biochemistry, Dongeui University College of Korean Medicine, 176 Yangjeong-ro, Busanjin-gu, Busan 47227, Republic of Korea
| |
Collapse
|
41
|
Lee MH, Cha HJ, Choi EO, Han MH, Kim SO, Kim GY, Hong SH, Park C, Moon SK, Jeong SJ, Jeong MJ, Kim WJ, Choi YH. Antioxidant and cytoprotective effects of morin against hydrogen peroxide-induced oxidative stress are associated with the induction of Nrf-2-mediated HO-1 expression in V79-4 Chinese hamster lung fibroblasts. Int J Mol Med 2017; 39:672-680. [DOI: 10.3892/ijmm.2017.2871] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 01/16/2017] [Indexed: 11/05/2022] Open
|
42
|
BitMansour A, Pop LM, Vitetta ES. The Role of Regulatory B Cell-Like Malignant Cells and Treg Cells in the Mouse Model of BCL1 Tumor Dormancy. PLoS One 2016; 11:e0167618. [PMID: 27959896 PMCID: PMC5154515 DOI: 10.1371/journal.pone.0167618] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 11/18/2016] [Indexed: 01/09/2023] Open
Abstract
Cancer dormancy is a clinical state in which residual tumor cells persist for long periods of time but do not cause detectable disease. In the mouse B cell lymphoma model (BCL1), dormancy can be induced and maintained by immunizing mice with a soluble form of the IgM expressed on the surface of the tumor cells. Immunization induces an anti-idiotype antibody response that maintains dormancy. Mice with dormant tumor have low numbers of BCL1 cells in their spleens that divide and are killed at the same rate. When the anti-Id antibodies wane, the tumor cells grow rapidly and kill the host. Spleens from tumor-bearing mice contain both effector (CD4+ and CD8+) and regulatory T cells (Tregs). In other tumor models, it has been reported that Tregs promote tumor progression by preventing effector cells from killing the tumor. In this report, we demonstrate that the tumor site with rapidly dividing BCL1 cells has fewer Tregs than the tumor site harboring dormant BCL1 cells. In both cases, the Tregs were equally suppressive in vitro. In spleens from mice with actively growing tumor, CD8+ but not CD4+ T cells were virtually absent. In vitro analysis demonstrated a tumor-mediated elimination of CD8+ T cells that was contact dependent and involved the caspase-3 pathway. Most importantly, we found that the BCL1 cells expressed characteristics of B10 regulatory B cells, i.e., they were CD1dhiCD5+ and secreted high levels of IL-10. These BCL1 tumor cells can inhibit anti-tumor immune responses by depleting CD8+ effector T cells.
Collapse
MESH Headings
- Animals
- Antibodies, Neoplasm/immunology
- Antigens, CD1/genetics
- Antigens, CD1/metabolism
- B-Lymphocytes, Regulatory/immunology
- B-Lymphocytes, Regulatory/pathology
- CD5 Antigens/genetics
- CD5 Antigens/metabolism
- Caspase 3/metabolism
- Cell Line
- Cells, Cultured
- Interleukin-10/genetics
- Interleukin-10/metabolism
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/pathology
- Mice
- Mice, Inbred BALB C
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
Collapse
Affiliation(s)
- Andrew BitMansour
- Department of Immunology and Cancer Immunobiology Center University of Texas Southwestern Medical Center at Dallas, Texas, United States of America
| | - Laurentiu M. Pop
- Department of Immunology and Cancer Immunobiology Center University of Texas Southwestern Medical Center at Dallas, Texas, United States of America
| | - Ellen S. Vitetta
- Department of Immunology and Cancer Immunobiology Center University of Texas Southwestern Medical Center at Dallas, Texas, United States of America
- * E-mail:
| |
Collapse
|
43
|
Chang YJ, Linh NH, Shih YH, Yu HM, Li MS, Chen YR. Alzheimer's Amyloid-β Sequesters Caspase-3 in Vitro via Its C-Terminal Tail. ACS Chem Neurosci 2016; 7:1097-106. [PMID: 27227450 DOI: 10.1021/acschemneuro.6b00049] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Amyloid-β (Aβ), the main constituent in senile plaques found in the brain of patients with Alzheimer's disease (AD), is considered as a causative factor in AD pathogenesis. The clinical examination of the brains of patients with AD has demonstrated that caspase-3 colocalizes with senile plaques. Cellular studies have shown that Aβ can induce neuronal apoptosis via caspase-3 activation. Here, we performed biochemical and in silico studies to investigate possible direct effect of Aβ on caspase-3 to understand the molecular mechanism of the interaction between Aβ and caspase-3. We found that Aβ conformers can specifically and directly sequester caspase-3 activity in which freshly prepared Aβ42 is the most potent. The inhibition is noncompetitive, and the C-terminal region of Aβ plays an important role in sequestration. The binding of Aβ to caspase-3 was examined by cross-linking and proteolysis and by docking and all-atom molecular dynamic simulations. Experimental and in silico results revealed that Aβ42 exhibits a higher binding affinity than Aβ40 and the hydrophobic C-terminal region plays a key role in the caspase-Aβ interaction. Overall, our study describes a novel mechanism demonstrating that Aβ sequesters caspase-3 activity via direct interaction and facilitates future therapeutic development in AD.
Collapse
Affiliation(s)
- Yu-Jen Chang
- Genomics
Research Center, Academia Sinica, Taiwan, 128, Academia Road, Sec. 2, Nankang
Dist., Taipei 115, Taiwan
| | - Nguyen Hoang Linh
- Institute for Computational Science and Technology, SBI Building,
Quang Trung Software City, Tan Chanh Hiep Ward, District 12, Ho Chi Minh City, Vietnam
| | - Yao Hsiang Shih
- Genomics
Research Center, Academia Sinica, Taiwan, 128, Academia Road, Sec. 2, Nankang
Dist., Taipei 115, Taiwan
| | - Hui-Ming Yu
- Genomics
Research Center, Academia Sinica, Taiwan, 128, Academia Road, Sec. 2, Nankang
Dist., Taipei 115, Taiwan
| | - Mai Suan Li
- Institute of Physics Polish Academy of Sciences, Al. Lotnikow 32/46, 02-668 Warsaw, Poland
| | - Yun-Ru Chen
- Genomics
Research Center, Academia Sinica, Taiwan, 128, Academia Road, Sec. 2, Nankang
Dist., Taipei 115, Taiwan
| |
Collapse
|
44
|
Choi EO, Park C, Hwang HJ, Hong SH, Kim GY, Cho EJ, Kim WJ, Choi YH. Baicalein induces apoptosis via ROS-dependent activation of caspases in human bladder cancer 5637 cells. Int J Oncol 2016; 49:1009-18. [PMID: 27571890 DOI: 10.3892/ijo.2016.3606] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 07/01/2016] [Indexed: 11/06/2022] Open
Abstract
Baicalein is a flavonoid derived originally from the root of Scutellaria baicalensis Georgi, which has been used in Oriental medicines for treating various diseases. Although this compound has been reported to have anticancer activities in several human cancer cell lines, the therapeutic effects of baicalein on human bladder cancer and its mechanisms of action have not been extensively studied. This study investigated the proapoptotic effects of baicalein in human bladder cancer 5637 cells. For this study, cell viability and apoptosis were evaluated using the 3-(4,5-dimethylthia-zol-2-yl)-2,5-diphenyltetrazolium bromide assay, trypan blue dye exclusion assay 4,6-diamidino-2-phenylindole staining, and flow cytometry. Measurements of the mitochondrial membrane potential (MMP), caspase activity assays and western blots were conducted to determine whether 5637 cell death occurred by apoptosis. Treatment with baicalein resulted in a concentration-dependent growth inhibition coupled with apoptosis induction, as indicated by the results of nuclei morphology examination and flow cytometry analyses. The induction of the apoptotic cell death of 5637 cells by baicalein exhibited a correlation with the downregulation of members of the inhibitor of apoptosis protein (IAP) family, including cIAP-1 and cIAP-2, and the activation of caspase-9 and -3 accompanied by proteolytic degradation of poly(ADP-ribose)-polymerase. The study also showed that baicalein decreases the expression of the proapoptotic protein Bax, increases antiapoptotic Bcl-2 expression, and noticeably aggravates the loss of MMP. Concomitantly, the data showed that baicalein increases the levels of death receptors and their associated ligands and enhances the activation of caspase-8 and truncation of Bid. However, the pan-caspase inhibitor can reverse baicalein-induced apoptosis, demonstrating that it is a caspase-dependent pathway. Moreover, it was found that baicalein can induce the production of reactive oxygen species (ROS) and that pretreatment with the antioxidant N-acetyl-L-cysteine significantly attenuates the baicalein effects on the loss of MMP and activation of caspase. In addition, the blocking of ROS generation decreases the apoptotic activity and antiproliferative effect of baicalein, indicating that baicalein induces apoptosis of 5637 cells through the ROS-dependent activation of caspases.
Collapse
Affiliation(s)
- Eun-Ok Choi
- Anti-Aging Research Center, Dongeui University, Busan 614-714, Republic of Korea
| | - Cheol Park
- Department of Molecular Biology, College of Natural Sciences and Human Ecology, Dongeui University, Busan 614-714, Republic of Korea
| | - Hye-Jin Hwang
- Anti-Aging Research Center, Dongeui University, Busan 614-714, Republic of Korea
| | - Su Hyun Hong
- Department of Biochemistry, Dongeui University College of Korean Medicine, Busan 614-052, Republic of Korea
| | - Gi-Young Kim
- Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National University, Jeju 690-756, Republic of Korea
| | - Eun-Ju Cho
- Department of Food and Nutrition, College of Human Ecology, Pusan National University, Busan 609-735, Republic of Korea
| | - Wun-Jae Kim
- Department of Urology, Chungbuk National University College of Medicine, Cheongju 361-763, Republic of Korea
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dongeui University, Busan 614-714, Republic of Korea
| |
Collapse
|
45
|
Lee TK, Park C, Jeong SJ, Jeong MJ, Kim GY, Kim WJ, Choi YH. Sanguinarine Induces Apoptosis of Human Oral Squamous Cell Carcinoma KB Cells via Inactivation of the PI3K/Akt Signaling Pathway. Drug Dev Res 2016; 77:227-40. [PMID: 27363951 DOI: 10.1002/ddr.21315] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 05/25/2016] [Indexed: 01/16/2023]
Abstract
Preclinical Research Sanguinarine, an alkaloid isolated from the root of Sanguinaria canadensis and other plants of the Papaveraceae family, selectively induces apoptotic cell death in a variety of human cancer cells, but its mechanism of action requires further elaboration. The present study investigated the pro-apoptotic effects of sanguinarine in human oral squamous cell carcinoma KB cells. Sanguinarine treatment increased DR5/TRAILR2 (death receptor 5/TRAIL receptor 2) expression and enhanced the activation of caspase-8 and cleavage of its substrate, Bid. Sanguinarine also induced the mitochondrial translocation of pro-apoptotic Bax, mitochondrial dysfunction, cytochrome c release to the cytosol, and activation of caspase-9 and -3. However, a pan-caspase inhibitor, z-VAD-fmk, reversed the growth inhibition and apoptosis induced by sanguinarine. Sanguinarine also suppressed the phosphorylation of phosphoinositide 3-kinase (PI3K) and Akt in KB cells, while co-treatment of cells with sanguinarine and a PI3K inhibitor revealed synergistic apoptotic effects. However, pharmacological inhibition of AMP-activated protein kinase and mitogen-activated protein kinases did not reduce or enhance sanguinarine-induced growth inhibition and apoptosis. Collectively, these findings indicate that the pro-apoptotic effects of sanguinarine in KB cells may be regulated by a caspase-dependent cascade via activation of both intrinsic and extrinsic signaling pathways and inactivation of PI3K/Akt signaling. Drug Dev Res 77 : 227-240, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tae Kyung Lee
- Department of Biology, New York University, New York, NY, 10012, USA
| | - Cheol Park
- Department of Molecular Biology, College of Natural Sciences and Human Ecology, Dongeui University, Busan, 614-714, South Korea
| | - Soon-Jeong Jeong
- Department of Dental Hygiene, College of Health Sciences, Youngsan University, Yangsan, 626-790, South Korea
| | - Moon-Jin Jeong
- Department of Oral Histology and Developmental Biology, School of Dentistry, Chosun University, Gwangju, 501-759, South Korea
| | - Gi-Young Kim
- Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National University, Jeju, 690-756, South Korea
| | - Wun-Jae Kim
- Department of Urology, Chungbuk National University, College of Medicine and Institute for Tumor Research, Cheongju, 28644, South Korea
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dongeui University, Busan, 614-714, South Korea.,Department of Biochemistry, Dongeui University College of Korean Medicine, Busan, 614-714, South Korea
| |
Collapse
|
46
|
Nagappan A, Lee HJ, Saralamma VVG, Park HS, Hong GE, Yumnam S, Raha S, Charles SN, Shin SC, Kim EH, Lee WS, Kim GS. Flavonoids isolated from Citrus platymamma induced G2/M cell cycle arrest and apoptosis in A549 human lung cancer cells. Oncol Lett 2016; 12:1394-1402. [PMID: 27446443 PMCID: PMC4950876 DOI: 10.3892/ol.2016.4793] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 04/29/2016] [Indexed: 12/28/2022] Open
Abstract
Citrus platymamma hort. ex Tanaka belongs to the Rutaceae family and is widely used in folk medicines in Korea due to its anti-proliferative, anti-cancer, anti-oxidant, anti-inflammatory and anti-diabetic activities. However, the molecular mechanism of its anti-cancer effect is not well understood. The present study was conducted to elucidate the anti-cancer effect and molecular mechanism of flavonoids from Citrus platymamma (FCP) on A549 cells. FCP displayed concentration-dependent inhibition on A549 cells proliferation. Further, flow cytometry revealed that FCP significantly increased the sub-G1 (apoptotic cell population) and G2/M phase population, and the total number of apoptotic cells, in a dose-dependent manner. Nuclear condensation and fragmentation were also observed upon staining with Hoechst 33342 in FCP-treated A549 cells. Immunoblotting demonstrated a dose-dependent downregulation of cyclin B1, cyclin-dependent kinase 1, cell division cycle 25c, pro-caspases −3, −6, −8 and −9, and poly (adenosine diphosphate-ribose) polymerase (PARP) in FCP-treated A549 cells. In addition, FCP induced caspase-3 activation and subsequent PARP cleavage, and increased the B-cell lymphoma (Bcl)-2-associated X protein/Bcl-extra large ratio in A549 cells. These findings suggest that FCP induced G2/M arrest and apoptosis of A549 cells. The present study provides evidence that FCP may be useful in the treatment of human lung cancer.
Collapse
Affiliation(s)
- Arulkumar Nagappan
- Department of Internal Medicine, Institute of Health Sciences and Gyeongnam Regional Cancer Center, School of Medicine, Gyeongsang National University, Jinju, Gyeongnam 660-702, Republic of Korea
| | - Ho Jeong Lee
- Brain Korea 21 Program for Leading Universities and Students, Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 660-701, Republic of Korea
| | - Venu Venkatarame Gowda Saralamma
- Brain Korea 21 Program for Leading Universities and Students, Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 660-701, Republic of Korea
| | - Hyeon Soo Park
- Brain Korea 21 Program for Leading Universities and Students, Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 660-701, Republic of Korea
| | - Gyeong Eun Hong
- Brain Korea 21 Program for Leading Universities and Students, Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 660-701, Republic of Korea
| | - Silvia Yumnam
- Brain Korea 21 Program for Leading Universities and Students, Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 660-701, Republic of Korea
| | - Suchismita Raha
- Brain Korea 21 Program for Leading Universities and Students, Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 660-701, Republic of Korea
| | - Shobana Nancy Charles
- Department of Internal Medicine, Institute of Health Sciences and Gyeongnam Regional Cancer Center, School of Medicine, Gyeongsang National University, Jinju, Gyeongnam 660-702, Republic of Korea
| | - Sung Chul Shin
- Department of Chemistry, Research Institute of Life Science, Gyeongsang National University, Jinju, Gyeongnam 660-701, Republic of Korea
| | - Eun Hee Kim
- Department of Nursing Science, International University of Korea, Jinju, Gyeongnam 660-759, Republic of Korea
| | - Won Sup Lee
- Department of Internal Medicine, Institute of Health Sciences and Gyeongnam Regional Cancer Center, School of Medicine, Gyeongsang National University, Jinju, Gyeongnam 660-702, Republic of Korea
| | - Gon Sup Kim
- Brain Korea 21 Program for Leading Universities and Students, Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 660-701, Republic of Korea
| |
Collapse
|
47
|
Yi J, Wang Z, Bai H, Li L, Zhao H, Cheng C, Zhang H, Li J. Polyphenols from pinecones of Pinus koraiensis induce apoptosis in colon cancer cells through the activation of caspase in vitro. RSC Adv 2016. [DOI: 10.1039/c5ra24913a] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The present study reports the antitumor effects of PPP-40 (the purified polyphenols from P. koraiensis pinecones by 40% ethanol) on LOVO cells and revealed its antitumor mechanism, which involved the apoptosis of cells associated with the activation of the caspase pathway.
Collapse
Affiliation(s)
- Juanjuan Yi
- School of Chemical Engineering
- Harbin Institute of Technology
- Harbin 150090
- PR China
| | - Zhenyu Wang
- School of Chemical Engineering
- Harbin Institute of Technology
- Harbin 150090
- PR China
| | - Haina Bai
- School of Chemical Engineering
- Harbin Institute of Technology
- Harbin 150090
- PR China
| | - Lu Li
- Northeast Agricultural University
- Harbin 150030
- PR China
| | - Haitian Zhao
- School of Chemical Engineering
- Harbin Institute of Technology
- Harbin 150090
- PR China
| | - Cuilin Cheng
- School of Chemical Engineering
- Harbin Institute of Technology
- Harbin 150090
- PR China
| | - Hua Zhang
- School of Chemical Engineering
- Harbin Institute of Technology
- Harbin 150090
- PR China
| | - Jingtong Li
- School of Chemical Engineering
- Harbin Institute of Technology
- Harbin 150090
- PR China
| |
Collapse
|
48
|
Silver nanoparticles provoke apoptosis of Dalton's ascites lymphoma in vivo by mitochondria dependent and independent pathways. Colloids Surf B Biointerfaces 2015; 136:1011-6. [PMID: 26590893 DOI: 10.1016/j.colsurfb.2015.11.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 10/31/2015] [Accepted: 11/02/2015] [Indexed: 02/07/2023]
Abstract
The aim of this report was to investigate the antitumor and apoptotic effects of silver nanoparticles (AgNPs) on the Dalton's ascites lymphoma cells in vivo. Thirty Swiss albino male mice were assigned into five groups of six each. Group I were intact animals. Group II animals served as tumor control injected with DAL cells intraperitonially. Group III induced animals received plant extract (17 mg/kg BW) and Group IV induced animals received AgNPs (35 μg/kg BW). Group V induced animals received standard anticancer drug 5-Fluorouracil (5-FU, 20 μg/kg BW). The treatment period was 10 days excluding the day of tumor injection. Tumor cells were collected after euthanizing the animals and real-time PCR was used to analyze p53, caspase-3, 8, 9, 12 and cytochrome C expressions. Results indicate that the AgNPs were efficient in prolongation of life span, reduction of tumor volume and body weight in tumor animals. All the apoptotic genes were upregulated by treatment with AgNPs. To conclude, the present study elicits that AgNPs are potent in antitumor activity and the molecular mechanism is by the induction of apoptosis through the mitochondrial dependent and independent pathways.
Collapse
|
49
|
Stefanowicz-Hajduk J, Bartoszewski R, Bartoszewska S, Kochan K, Adamska A, Kosiński I, Ochocka JR. Pennogenyl Saponins from Paris quadrifolia L. Induce Extrinsic and Intrinsic Pathway of Apoptosis in Human Cervical Cancer HeLa Cells. PLoS One 2015; 10:e0135993. [PMID: 26295969 PMCID: PMC4546673 DOI: 10.1371/journal.pone.0135993] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/28/2015] [Indexed: 11/19/2022] Open
Abstract
Pennogenyl saponins are the active compounds of large number of plant species and consequently many polyherbal formulations. Hence, great interest has been shown in their characterization and in the investigation of their pharmacological and biological properties, especially anticancer. This present study reports on the evaluation of cytotoxic effects and explanation of the molecular mechanisms of action of the two pennogenyl saponins (PS 1 and PS 2) isolated from Paris quadrifolia L. rhizomes on human cervical adenocarcinoma cell line HeLa. To determine the viability of the cells treated with the compounds we used real-time cell proliferation analysis and found that the pennogenyl saponins PS 1 and PS 2 strongly inhibited the tumor cells growth with IC50 values of 1.11 ± 0.04 μg/ml and 0.87 ± 0.05 μg/ml, respectively. The flow cytometry analysis indicated that the two compounds induced apoptosis in a dose-dependent manner and decreased mitochondrial membrane potential in HeLa cells in the early stage of apoptosis. Quantitative PCR and Western Blot analysis showed that the two saponins significantly increased mRNA expression of FADD and BID as well as induced caspase-8 via increased of procaspase-8 processing in the treated cells. The results of this study suggest that both the extrinsic death receptor and intrinsic mitochondrial pathways are involved in the programmed cell death.
Collapse
Affiliation(s)
| | - Rafal Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Sylwia Bartoszewska
- Department of Inorganic Chemistry, Medical University of Gdansk, Gdansk, Poland
| | - Kinga Kochan
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Anna Adamska
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Igor Kosiński
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - J. Renata Ochocka
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
50
|
Yoon JM, Koppula S, Huh SJ, Hur SJ, Kim CG. Low concentrations of doxycycline attenuates FasL-induced apoptosis in HeLa cells. Biol Res 2015. [PMID: 26205793 PMCID: PMC4511997 DOI: 10.1186/s40659-015-0025-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background Doxycycline (DC) has been shown to possess non-antibiotic properties including Fas/Fas Ligand (FasL)-mediated apoptosis against several tumor types in the concentration range of 10–40 µg/mL. However, the effect of DC in apoptotic signaling at much low concentrations was not studied. Methods The present study investigated the attenuation effect of low dose of DC on FasL-induced apoptosis in HeLa cell by the methods of MTT assay, fluorescence microscopy, DNA fragmentation, flow cytometry analysis, and western blotting. Results and conclusion In the present findings we showed that low concentration of DC (<2.0 µg/mL) exhibited protective effects against FasL-induced apoptosis in HeLa cells. FasL treatment to HeLa cells resulted in a concentration-dependent induction of cell death, and treatment with low concentrations of DC (0.1–2 µg/mL) significantly (p < 0.001) attenuated the FasL-induced cell death as measured by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Further, the FasL-induced apoptotic features in HeLa cells, such as morphological changes, DNA fragmentation and cell cycle arrest was also inhibited by DC (0.5 µg/mL). Tetracycline and minocycline also showed similar anti-apoptotic effects but were not significant when compared to DC, tested at same concentrations. Further, DC (0.01–16 µg/mL) did not influence the hydrogen peroxide- or cisplatin-induced intrinsic apoptotic pathway in HeLa cells. Protein analysis using Western blotting confirmed that FasL-induced cleavage/activation of caspase-8 and caspase-3, were inhibited by DC treatment at low concentration (0.5 µg/mL). Considering the overall data, we report for the first time that DC exhibited anti-apoptotic effects at low concentrations in HeLa cells by inhibition of caspase activation via FasL-induced extrinsic pathway. Electronic supplementary material The online version of this article (doi:10.1186/s40659-015-0025-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jung Mi Yoon
- Department of Biotechnology, Konkuk University, Chungju, 380-701, Republic of Korea.
| | - Sushruta Koppula
- Department of Biotechnology, Konkuk University, Chungju, 380-701, Republic of Korea.
| | - Se Jong Huh
- Department of Biotechnology, Konkuk University, Chungju, 380-701, Republic of Korea.
| | - Sun Jin Hur
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Gyeonggi, 456-756, South Korea.
| | - Chan Gil Kim
- Department of Biotechnology, Konkuk University, Chungju, 380-701, Republic of Korea.
| |
Collapse
|