1
|
Yuce M, Albayrak E. Paracrine Factors Released from Tonsil-Derived Mesenchymal Stem Cells Inhibit Proliferation of Hematological Cancer Cells Under Hyperthermia in Co-culture Model. Appl Biochem Biotechnol 2024; 196:4105-4124. [PMID: 37897623 DOI: 10.1007/s12010-023-04757-7] [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] [Accepted: 10/17/2023] [Indexed: 10/30/2023]
Abstract
Mesenchymal stem cells (MSCs) are promising biological therapeutic candidates in cancer treatment. As a source of MSCs, palatine tonsil tissue is one of the secondary lymphoid organs that form an essential part of the immune system, and the relation between the secondary lymphoid organs and cancer progression leads us to investigate the effect of tonsil-derived MSCs (T-MSC) on cancer treatment. We aimed to determine the anti-tumoral effects of T-MSCs cultured at the febrile temperature (40 °C) on hematological cancer cell lines. The co-culture of cancer cells with T-MSCs was carried out under fever and normal culture conditions, and then the cell viability was determined by cell counting. In addition, apoptosis rate and cell cycle arrest were determined by flow cytometry. We confirmed the apoptotic effect of T-MSC co-culture at the transcriptional level by using real-time polymerase chain reaction (RT-PCR). We found that co-culture of cancer cells with T-MSCs significantly decreased the viable cell number under the febrile and normal culture conditions. Besides, the T-MSC co-culture induced apoptosis on K562 and MOLT-4 cells and induced the cell cycle arrest at the G2/M phase on MOLT-4 cells. The apoptotic effect of T-MSC co-culture under febrile stimulation was confirmed at the transcriptional level. Our study has highlighted the anti-tumoral effect of the cellular interaction between the T-MSCs and human hematological cancer cells during in vitro co-culture under hyperthermia.
Collapse
Affiliation(s)
- Melek Yuce
- Stem Cell Research & Application Center, Ondokuz Mayıs University, Kurupelit Campus, 55139, Atakum, Samsun, Turkey.
| | - Esra Albayrak
- Stem Cell Research & Application Center, Ondokuz Mayıs University, Kurupelit Campus, 55139, Atakum, Samsun, Turkey
| |
Collapse
|
2
|
Haneen DSA, Hekal MH, Abou-Elmagd WSI, El-Sayed WM. Novel pyrano[2,3-c]pyrazolopyrimidines as promising anticancer agents: Design, synthesis, and cell cycle arrest of HepG2 cells at S phase. SYNTHETIC COMMUN 2024; 54:655-671. [DOI: 10.1080/00397911.2024.2327047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Indexed: 10/06/2024]
Affiliation(s)
- David S. A. Haneen
- Department of Chemistry, Faculty of Science, Ain Shams University, Abbassia, Cairo, Egypt
| | - Mohamed H. Hekal
- Department of Chemistry, Faculty of Science, Ain Shams University, Abbassia, Cairo, Egypt
| | - Wael S. I. Abou-Elmagd
- Department of Chemistry, Faculty of Science, Ain Shams University, Abbassia, Cairo, Egypt
| | - Wael M. El-Sayed
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, Cairo, Egypt
| |
Collapse
|
3
|
Nguyen C, Toubia I, Hadj-Kaddour K, Ali LMA, Lichon L, Cure C, Diring S, Kobeissi M, Odobel F, Gary-Bobo M. Exceptional anticancer photodynamic properties of [1,4-Bis(3,6,9,12-Tetraoxatridec-1-yloxy)phthalocyaninato]zinc(II). JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 253:112863. [PMID: 38457992 DOI: 10.1016/j.jphotobiol.2024.112863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/18/2024] [Accepted: 02/05/2024] [Indexed: 03/10/2024]
Abstract
Phthalocyanines have been described as effective photosensitizers for photodynamic therapy and are therefore, being studied for their biomedical applications. The metalation of photosensitizers can improve their photodynamic therapy potential. Here, we focus on the biological properties of [1,4-Bis(3,6,9,12-Tetraoxatridec-1-yloxy)phthalocyaninato]zinc(II) (ZnPc(αEG4)2) and demonstrate its exceptional anticancer activity upon light stimulation to kill preferentially cancer cells with a start of efficiency at 10 pM. Indeed, in this work we highlighted the high selectivity of ZnPc(αEG4)2 for cancer cells compared with healthy ones and we establish its mechanism of action, enabling us to conclude that ZnPc(αEG4)2 could be a powerful tool for cancer therapy.
Collapse
Affiliation(s)
| | - Isabelle Toubia
- Nantes Université, CNRS, CEISAM, UMR 6230, F-44000 Nantes, France
| | | | - Lamiaa M A Ali
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France; Department of Biochemistry, Medical Research Institute, University of Alexandria, 21561 Alexandria, Egypt
| | - Laure Lichon
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Charlotte Cure
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Stéphane Diring
- Nantes Université, CNRS, CEISAM, UMR 6230, F-44000 Nantes, France
| | - Marwan Kobeissi
- Laboratoire RammalRammal, Equipe de Synthèse Organique Appliquée SOA, Université Libanaise, Faculté des Sciences 5, Nabatieh, Lebanon.
| | - Fabrice Odobel
- Nantes Université, CNRS, CEISAM, UMR 6230, F-44000 Nantes, France.
| | | |
Collapse
|
4
|
Ho YF, Yajit NLM, Shiau JY, Malek SNA, Shyur LF, Karsani SA. Changes in the Proteome Profile of A549 Cells Following Helichrysetin-Induced Apoptosis Suggest the Involvement of DNA Damage Response and Cell Cycle Arrest-Associated Proteins. Appl Biochem Biotechnol 2023; 195:6867-6880. [PMID: 36947367 DOI: 10.1007/s12010-023-04384-2] [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] [Accepted: 02/17/2023] [Indexed: 03/23/2023]
Abstract
Our previous findings demonstrated that Helichrysetin possessed promising anti-cancer activity. It was able to induce apoptosis in the A549 cell line. However, its mechanism of action is unknown. The present study aimed to unravel possible underlying molecular mechanisms of helichrysetin-induced apoptosis in A549 (human lung carcinoma) cells using comparative quantitative proteomics (iTRAQ labeled), followed by an exhaustive bioinformatics analysis. Our results suggested that DNA damage response (DDR) and cell cycle arrest were responsible for lung cancer cell death with helichrysetin treatment. Among proteins that changed in abundance were Nrf2 and HMOX1. They are oxidative stress-related proteins and were increased in abundance. BRAT1 was also increased in abundance, suggesting an increase in DNA damage repair, indicating the occurrence of DNA damage due to oxidative stress. However, several essential DDR downstream proteins such as p-ATM, BRCA1, FANCD2, and Rb1 that would further increase DNA damage were found to be dramatically decreased in relative abundance. Cell cycle-related proteins, p53, p21, and cyclin D1, were increased while cyclin A, cyclin E, and cdk2 were decreased. This is predicted to facilitate S-phase arrest. Furthermore, excessive DNA damage and prolonged arrest would in turn result in the induction of mitochondrial-mediated apoptosis. Based on these observations, we postulate that the effects of helichrysetin were in part via the suppression of DNA damage response which led to DNA damage and prolonged cell cycle arrest. Subsequently, this event initiated mitochondrial-mediated apoptosis in A549 lung cancer cells.
Collapse
Affiliation(s)
- Yen Fong Ho
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Noor Liana Mat Yajit
- University of Malaya Centre for Proteomics Research (UMCPR), Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Jeng-Yuan Shiau
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, 115, Taiwan
- Institute of Biotechnology, National Taiwan University, Taipei, 106, Taiwan
| | - Sri Nurestri Abd Malek
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Lie-Fen Shyur
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, 115, Taiwan.
- Institute of Biotechnology, National Taiwan University, Taipei, 106, Taiwan.
| | - Saiful Anuar Karsani
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
5
|
Wei L, Zhang Q, Zhong C, He L, Zhang Y, Armaly AM, Aubé J, Welch DR, Xu L, Wu X. Functional inhibition of the RNA-binding protein HuR sensitizes triple-negative breast cancer to chemotherapy. Mol Oncol 2023; 17:1962-1980. [PMID: 37357618 PMCID: PMC10552894 DOI: 10.1002/1878-0261.13478] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/18/2023] [Accepted: 06/23/2023] [Indexed: 06/27/2023] Open
Abstract
Chemotherapy remains the standard treatment for triple-negative breast cancer (TNBC); however, chemoresistance compromises its efficacy. The RNA-binding protein Hu antigen R (HuR) could be a potential therapeutic target to enhance the chemotherapy efficacy. HuR is known to mainly stabilize its target mRNAs, and/or promote the translation of encoded proteins, which are implicated in multiple cancer hallmarks, including chemoresistance. In this study, a docetaxel-resistant cell subline (231-TR) was established from the human TNBC cell line MDA-MB-231. Both the parental and resistant cell lines exhibited similar sensitivity to the small molecule functional inhibitor of HuR, KH-3. Docetaxel and KH-3 combination therapy synergistically inhibited cell proliferation in TNBC cells and tumor growth in three animal models. KH-3 downregulated the expression levels of HuR targets (e.g., β-Catenin and BCL2) in a time- and dose-dependent manner. Moreover, KH-3 restored docetaxel's effects on activating Caspase-3 and cleaving PARP in 231-TR cells, induced apoptotic cell death, and caused S-phase cell cycle arrest. Together, our findings suggest that HuR is a critical mediator of docetaxel resistance and provide a rationale for combining HuR inhibitors and chemotherapeutic agents to enhance chemotherapy efficacy.
Collapse
Affiliation(s)
- Lanjing Wei
- Bioengineering ProgramThe University of KansasLawrenceKSUSA
| | - Qi Zhang
- Department of Molecular BiosciencesThe University of KansasLawrenceKSUSA
| | - Cuncong Zhong
- Department of Electrical Engineering and Computer ScienceThe University of KansasLawrenceKSUSA
| | - Lily He
- Department of Pharmacology, Toxicology & TherapeuticsThe University of Kansas Medical CenterKansas CityKSUSA
| | - Yuxia Zhang
- Department of Pharmacology, Toxicology & TherapeuticsThe University of Kansas Medical CenterKansas CityKSUSA
| | - Ahlam M. Armaly
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of PharmacyThe University of North CarolinaChapel HillNCUSA
| | - Jeffrey Aubé
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of PharmacyThe University of North CarolinaChapel HillNCUSA
| | - Danny R. Welch
- Department of Cancer BiologyThe University of Kansas Medical CenterKansas CityKSUSA
- The University of Kansas Cancer CenterThe University of Kansas Medical CenterKansas CityKSUSA
| | - Liang Xu
- Department of Molecular BiosciencesThe University of KansasLawrenceKSUSA
- The University of Kansas Cancer CenterThe University of Kansas Medical CenterKansas CityKSUSA
- Department of Radiation OncologyThe University of Kansas Medical CenterKansas CityKSUSA
| | - Xiaoqing Wu
- The University of Kansas Cancer CenterThe University of Kansas Medical CenterKansas CityKSUSA
- Higuchi Biosciences CenterThe University of KansasLawrenceKSUSA
| |
Collapse
|
6
|
Rodak O, Mrozowska M, Rusak A, Gomułkiewicz A, Piotrowska A, Olbromski M, Podhorska-Okołów M, Ugorski M, Dzięgiel P. Targeting SOX18 Transcription Factor Activity by Small-Molecule Inhibitor Sm4 in Non-Small Lung Cancer Cell Lines. Int J Mol Sci 2023; 24:11316. [PMID: 37511076 PMCID: PMC10379584 DOI: 10.3390/ijms241411316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/22/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
The transcription factor SOX18 has been shown to play a crucial role in lung cancer progression and metastasis. In this study, we investigated the effect of Sm4, a SOX18 inhibitor, on cell cycle regulation in non-small cell lung cancer (NSCLC) cell lines LXF-289 and SK-MES-1, as well as normal human lung fibroblast cell line IMR-90. Our results demonstrated that Sm4 treatment induced cytotoxic effects on all three cell lines, with a greater effect observed in NSCLC adenocarcinoma cells. Sm4 treatment led to S-phase cell accumulation and upregulation of p21, a key regulator of the S-to-G2/M phase transition. While no significant changes in SOX7 or SOX17 protein expression were observed, Sm4 treatment resulted in a significant upregulation of SOX17 gene expression. Furthermore, our findings suggest a complex interplay between SOX18 and p21 in the context of lung cancer, with a positive correlation observed between SOX18 expression and p21 nuclear presence in clinical tissue samples obtained from lung cancer patients. These results suggest that Sm4 has the potential to disrupt the cell cycle and target cancer cell growth by modulating SOX18 activity and p21 expression. Further investigation is necessary to fully understand the relationship between SOX18 and p21 in lung cancer and to explore the therapeutic potential of SOX18 inhibition in lung cancer.
Collapse
Affiliation(s)
- Olga Rodak
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Monika Mrozowska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Agnieszka Rusak
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Agnieszka Gomułkiewicz
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Aleksandra Piotrowska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Mateusz Olbromski
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Marzenna Podhorska-Okołów
- Division of Ultrastructural Research, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Maciej Ugorski
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Department of Physiotherapy, University School of Physical Education, 51-612 Wroclaw, Poland
| |
Collapse
|
7
|
Novinbahador T, Araj-Khodaei M, Mahdavi M. Evidence for Hesperidin as an Effective Factor in Initiating the Intrinsic Pathway of Apoptosis in KG1a Leukemia Cells. Int J Toxicol 2023; 42:165-171. [PMID: 36534417 DOI: 10.1177/10915818221146468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Acute myeloid leukemia (AML) is the most common subtype of leukemia, accounting for 62% of all leukemia fatalities. As a polyphenol glycoside, hesperidin triggers the apoptotic pathway, which might positively affect combating cancer cells. In this study, we investigated the pro-apoptotic effects of hesperidin in KG1a cells. The MTT assay was used to determine the IC50 of hesperidin in KG1a cell lines. For the apoptotic cell morphology study, we used Hoechst 33 258 staining. Activation of the caspase-3 enzyme was evaluated by the caspase-3 assay and spectrophotometry. Cell cycle distribution was analyzed by propidium iodide staining and flow cytometry. Moreover, p21, survivin, Bax, and Bcl2 gene expression was investigated by real-time PCR. Hesperidin decreased the viability of KG1a leukemic cell4s, but not that of HFF2, a non-cancer cell line. Apoptotic cell morphological alterations and increase in caspase-3 activity were observed after hesperidin treatment. Our results revealed that the expression of anti-apoptotic genes survivin and Bcl2 significantly decreased with hesperidin treatment, and pro-apoptotic gene Bax and cell cycle regulator p21 increased compared to the control group. These findings revealed that hesperidin may be an effective factor in initiating the intrinsic pathway of apoptosis and may be good candidate for the treatment of AML.
Collapse
Affiliation(s)
- Tannaz Novinbahador
- Department of Biology, Faculty of Natural Sciences, 56947University of Tabriz, Tabriz, Iran
| | - Mostafa Araj-Khodaei
- Department of Persian Medicine, Faculty of Traditional Medicine, 48432Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Mahdavi
- Department of Biology, Faculty of Natural Sciences, 56947University of Tabriz, Tabriz, Iran.,Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| |
Collapse
|
8
|
Turner BRH, Mellor C, McElroy C, Bowen N, Gu W, Knill C, Itasaki N. Non-ubiquitous expression of core spliceosomal protein SmB/B' in chick and mouse embryos. Dev Dyn 2023; 252:276-293. [PMID: 36058892 PMCID: PMC10087933 DOI: 10.1002/dvdy.537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/02/2022] [Accepted: 08/25/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Although splicing is an integral part of the expression of many genes in our body, genetic syndromes with spliceosomal defects affect only specific tissues. To help understand the mechanism, we investigated the expression pattern of a core protein of the major spliceosome, SmB/B' (Small Nuclear Ribonucleoprotein Polypeptides B/B'), which is encoded by SNRPB. Loss-of-function mutations of SNRPB in humans cause cerebro-costo-mandibular syndrome (CCMS) characterized by rib gaps, micrognathia, cleft palate, and scoliosis. Our expression analysis focused on the affected structures as well as non-affected tissues, using chick and mouse embryos as model animals. RESULTS Embryos at young stages (gastrula) showed ubiquitous expression of SmB/B'. However, the level and pattern of expression became tissue-specific as differentiation proceeded. The regions relating to CCMS phenotypes such as cartilages of ribs and vertebrae and palatal mesenchyme express SmB/B' in the nucleus sporadically. However, cartilages that are not affected in CCMS also showed similar expressions. Another spliceosomal gene, SNRNP200, which mutations cause retinitis pigmentosa, was also prominently expressed in cartilages in addition to the retina. CONCLUSION The expression of SmB/B' is spatiotemporally regulated during embryogenesis despite the ubiquitous requirement of the spliceosome, however, the expression pattern is not strictly correlated with the phenotype presentation.
Collapse
Affiliation(s)
| | | | - Clara McElroy
- Faculty of Health Sciences, University of Bristol, Bristol, UK
| | - Natalie Bowen
- Faculty of Life Sciences, University of Bristol, Bristol, UK
| | - Wenjia Gu
- Faculty of Life Sciences, University of Bristol, Bristol, UK
| | - Chris Knill
- Faculty of Life Sciences, University of Bristol, Bristol, UK
| | - Nobue Itasaki
- Faculty of Health Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
9
|
Lee H, An G, Lim W, Song G. Pendimethalin exposure induces bovine mammary epithelial cell death through excessive ROS production and alterations in the PI3K and MAPK signaling pathways. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2022; 188:105254. [PMID: 36464334 DOI: 10.1016/j.pestbp.2022.105254] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/13/2022] [Accepted: 09/20/2022] [Indexed: 06/17/2023]
Abstract
Herbicides are chemicals that have been established to have adverse impacts. However, they are still widely used in agriculture. Pendimethalin (PDM) is an herbicide that is widely used in many countries to control annual grasses. The possibility of livestock being exposed to PDM is relatively high, considering the half-life of PDM and its residues in water, soil and crops. However, the toxicity of PDM in cattle, especially in the mammary glands, has not been reported. Therefore, we investigated whether PDM has toxic effects in the mammary epithelial cells (MAC-T) of cattle. MAC-T cells were treated with various doses (0, 2.5, 5 and 10 μM) of PDM. We found that PDM affected cell viability and cell proliferation and causes cell cycle arrest. Furthermore, PDM triggered cell apoptosis, induced excessive ROS production and mitochondrial membrane potential (MMP) loss, and disrupted calcium homeostasis. In addition, PDM altered the activation of proteins associated with the endoplasmic reticulum (ER) stress response and modified PI3K and MAPK signaling cascades. In conclusion, our current study unveiled the mechanism of PDM in MAC-T cells and we suggest that PDM might be harmful to the mammary gland system of cattle, possibly affecting milk production.
Collapse
Affiliation(s)
- Hojun Lee
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Garam An
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Whasun Lim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
10
|
Taxifolin and Lucidin as Potential E6 Protein Inhibitors: p53 Function Re-Establishment and Apoptosis Induction in Cervical Cancer Cells. Cancers (Basel) 2022; 14:cancers14122834. [PMID: 35740499 PMCID: PMC9221127 DOI: 10.3390/cancers14122834] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 05/29/2022] [Accepted: 06/06/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Human papillomavirus (HPV)-related cancers continue to be a major medical concern, and there exists an urgent need to improve the current therapeutic approaches by combining strategies or proposing new compounds to offer more specific and less invasive treatments. The aim of this work was to discover potential inhibitors of the E6/E6AP/p53 complex formation. We started this work with an initial in silico approach including molecular docking and molecular dynamics simulations, and these tools allowed us to select potential inhibitors, using E6 protein as a target. In addition, we found that lucidin and taxifolin were able to selectively decrease the viability of HPV-positive cells to re-establish p53 protein levels and to induce apoptosis. These findings represent a promising starting point for the development of anti-HPV drugs. Abstract Cervical cancer is the fourth leading cause of death in women worldwide, with 99% of cases associated with a human papillomavirus (HPV) infection. Given that HPV prophylactic vaccines do not exert a therapeutic effect in individuals previously infected, have low coverage of all HPV types, and have poor accessibility in developing countries, it is unlikely that HPV-associated cancers will be eradicated in the coming years. Therefore, there is an emerging need for the development of anti-HPV drugs. Considering HPV E6’s oncogenic role, this protein has been proposed as a relevant target for cancer treatment. In the present work, we employed in silico tools to discover potential E6 inhibitors, as well as biochemical and cellular assays to understand the action of selected compounds in HPV-positive cells (Caski and HeLa) vs. HPV-negative (C33A) and non-carcinogenic (NHEK) cell lines. In fact, by molecular docking and molecular dynamics simulations, we found three phenolic compounds able to dock in the E6AP binding pocket of the E6 protein. In particular, lucidin and taxifolin were able to inhibit E6-mediated p53 degradation, selectively reduce the viability, and induce apoptosis in HPV-positive cells. Altogether, our data can be relevant for discovering promising leads for the development of specific anti-HPV drugs.
Collapse
|
11
|
Yang Z, Xie J, Fang J, Lv M, Yang M, Deng Z, Xie Y, Cai L. Nigericin exerts anticancer effects through inhibition of the SRC/STAT3/BCL-2 in osteosarcoma. Biochem Pharmacol 2022; 198:114938. [PMID: 35114189 DOI: 10.1016/j.bcp.2022.114938] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 11/02/2022]
Abstract
The treatment of osteosarcoma has reached a bottleneck period in recent 30 years, there is an urgent need to find new drugs and treatment methods. Nigericin, an antibiotic derived from Streptomyces hygroscopicus, has exerted promising antitumoral effect in various tumors. The anticancer effect of Nigericin in human osteosarcoma has never been reported. In the present study, we explored the anticancer effects of Nigericin in osteosarcoma in vitro and in vivo. Our results showed that nigericin treatment significantly reduced tumor cell proliferation in dose-dependent and time-dependent in human osteosarcoma cells. Nigericin can inhibit cell growth of osteosarcoma cells, in addition to S-phase cycle arrest, the nigericin induces apoptosis. Furthermore, bioinformatics predicted that Nigericin exerts anticancer effects through inhibiting SRC/STAT3 signaling pathway in osteosarcoma. The direct binding between SRC and activator of transcription 3 (STAT3) was confirmed by Western blot. Nigericin can down regulate STAT3 and Bcl-2. In order to further elucidate the inhibitory effect of nigericin on SRC / STAT3 / Bcl-2 signal transduction mechanism, we established human osteosarcoma cancer cells stably expressing STAT3. Western blot confirmed that nigericin exerts anticancer effects on human osteosarcoma cancer cells by directly targeting STAT3. In addition, Nigericin can significantly inhibit tumor migration and invasion. Finally, Nigericin inhibits tumor growth in a mouse osteosarcoma model. The nigericin targeting the SRC/STAT3/BCL-2 signaling pathway may provide new insights into the molecular mechanism of nigericin on cancer cells and suggest its possible clinical application in osteosarcoma.
Collapse
Affiliation(s)
- Zhiqiang Yang
- The Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, People's Republic of China.
| | - Jiangtao Xie
- The Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, People's Republic of China.
| | - Jiayu Fang
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430000, China.
| | - Minchao Lv
- The Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, People's Republic of China.
| | - Min Yang
- The Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, People's Republic of China.
| | - Zhouming Deng
- The Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, People's Republic of China.
| | - Yuanlong Xie
- The Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, People's Republic of China.
| | - Lin Cai
- The Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, People's Republic of China.
| |
Collapse
|
12
|
Kim IH, Eom T, Park JY, Kim HJ, Nam TJ. Dichloromethane fractions of Calystegia soldanella induce S‑phase arrest and apoptosis in HT‑29 human colorectal cancer cells. Mol Med Rep 2021; 25:60. [PMID: 34935054 PMCID: PMC8767546 DOI: 10.3892/mmr.2021.12576] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/11/2021] [Indexed: 11/22/2022] Open
Abstract
Calystegia soldanella is a halophyte and a perennial herb that grows on coastal sand dunes worldwide. Extracts from this plant have been previously revealed to have a variety of bioactive properties in humans. However, their effects on colorectal cancer cells remain poorly understood. In the present study, the potential biological activity of C. soldanella extracts in the colorectal cancer cell line HT-29 was examined. First, five solvent fractions [n-hexane, dichloromethane (DCM), ethyl acetate, n-butanol and water] were obtained from the crude extracts of C. soldanella through an organic solvent extraction method. In particular, the DCM fraction was demonstrated to exert marked dose- and time-dependent inhibitory effects according to results from the cell viability assay. Data obtained from the apoptosis assay suggested that the inhibition of HT-29 cell viability induced by DCM treatment was attributed to increased apoptosis. The apoptotic rate was markedly increased in a dose-dependent manner, which was associated with the protein expression levels of apoptosis-related proteins, including increased Fas, Bad and Bax, and decreased pro-caspase-8, Bcl-2, Bcl-xL, pro-caspase-9, pro-caspase-7 and pro-caspase-3. A mitochondrial membrane potential assay demonstrated that more cells became depolarized and the extent of cytochrome c release was markedly increased in a dose-dependent manner in HT-29 cells treated with DCM. In addition, cell cycle analysis confirmed S-phase arrest following DCM fraction treatment, which was associated with decreased protein expression levels of cell cycle-related proteins, such as cyclin A, CDK2, cell division cycle 25 A and cyclin dependent kinase inhibitor 1. Based on these results, the present study suggested that the DCM fraction of the C. soldanella extract can inhibit HT-29 cell viability whilst inducing apoptosis through mitochondrial membrane potential regulation and S-phase arrest. These results also suggested that the DCM fraction has potential anticancer activity in HT-29 colorectal cells. Further research on the composition of the DCM fraction is warranted.
Collapse
Affiliation(s)
- In-Hye Kim
- Future Fisheries Food Research Center, Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Republic of Korea
| | - Taekil Eom
- Future Fisheries Food Research Center, Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Republic of Korea
| | - Joon-Young Park
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea
| | - Hyung-Joo Kim
- Future Fisheries Food Research Center, Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Republic of Korea
| | - Taek-Jeong Nam
- Future Fisheries Food Research Center, Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Republic of Korea
| |
Collapse
|
13
|
Maharjan S, Kwon YS, Lee MG, Lee KS, Nam KS. Cell cycle arrest-mediated cell death by morin in MDA-MB-231 triple-negative breast cancer cells. Pharmacol Rep 2021; 73:1315-1327. [PMID: 33993438 DOI: 10.1007/s43440-021-00272-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Morin, a flavonoid extracted from Moraceace family and exhibits several pharmacological activities including anti-cancer activity. Although the anticancer activity of morin in breast cancer was estimated in some investigations, the pharmaceutical mechanism has not been fully elucidated. Therefore, we investigated to unveil the detail signaling pathway in morin-treated in MDA-MB-231 triple-negative breast cancer cells. METHODS The cytotoxicity of morin in MDA-MB-231 cells was confirmed by sulforhodamine B (SRB) assay and colony formation assay. Flow cytometry was performed to examine the cell cycle and cell death patterns and the protein expression and phosphorylation were detected by western blotting. RESULTS Our results showed that morin inhibited MDA-MB-231 cells proliferation in time and concentration-dependent manner. Morphological changes were observed when treated with various concentration of morin in MDA-MB-231 cells. In regard to protein expression, morin induced the phosphorylation of ERK and p-H2A.X and decreased the level of DNA repair markers, RAD51 and survivin. In addition, flow cytometry showed S and G2/M arrest by morin that was associated with the decrease in the protein expression of cyclin A2 and cyclin B1 and upregulation of p21. Interestingly, annexin V/PI staining result clearly showed that morin induced cell death without apoptosis. Furthermore, attenuated FoxM1 by morin was co-related with cell cycle regulators including p21, cyclin A2 and cyclin B1. CONCLUSION Taken together, our study indicates that morin-induced cell death of MDA-MB-231 is caused by sustained cell cycle arrest via the induction of p21 expression by activation of ERK and repression of FOXM1 signaling pathways.
Collapse
Affiliation(s)
- Sushma Maharjan
- Department of Pharmacology, College of Medicine and Intractable Disease Research Center, Dongguk University, Gyeongju, 38066, Republic of Korea
| | - Yun-Suk Kwon
- Department of Pharmacology, College of Medicine and Intractable Disease Research Center, Dongguk University, Gyeongju, 38066, Republic of Korea
| | - Min-Gu Lee
- Department of Pharmacology, College of Medicine and Intractable Disease Research Center, Dongguk University, Gyeongju, 38066, Republic of Korea
| | - Kyu-Shik Lee
- Department of Pharmacology, College of Medicine and Intractable Disease Research Center, Dongguk University, Gyeongju, 38066, Republic of Korea
| | - Kyung-Soo Nam
- Department of Pharmacology, College of Medicine and Intractable Disease Research Center, Dongguk University, Gyeongju, 38066, Republic of Korea.
| |
Collapse
|
14
|
Wu PS, Yen JH, Wang CY, Chen PY, Hung JH, Wu MJ. 8-Hydroxydaidzein, an Isoflavone from Fermented Soybean, Induces Autophagy, Apoptosis, Differentiation, and Degradation of Oncoprotein BCR-ABL in K562 Cells. Biomedicines 2020; 8:E506. [PMID: 33207739 PMCID: PMC7696406 DOI: 10.3390/biomedicines8110506] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 12/22/2022] Open
Abstract
8-Hydroxydaidzein (8-OHD, 7,8,4'-trihydoxyisoflavone) is a hydroxylated derivative of daidzein isolated from fermented soybean products. The aim of this study is to investigate the anti-proliferative effects and the underlying mechanisms of 8-OHD in K562 human chronic myeloid leukemia (CML) cells. We found that 8-OHD induced reactive oxygen species (ROS) overproduction and cell cycle arrest at the S phase by upregulating p21Cip1 and downregulating cyclin D2 (CCND2) and cyclin-dependent kinase 6 (CDK6) expression. 8-OHD also induced autophagy, caspase-7-dependent apoptosis, and the degradation of BCR-ABL oncoprotein. 8-OHD promoted Early Growth Response 1 (EGR1)-mediated megakaryocytic differentiation as an increased expression of marker genes, CD61 and CD42b, and the formation of multi-lobulated nuclei in enlarged K562 cells. A microarray-based transcriptome analysis revealed a total of 3174 differentially expressed genes (DEGs) after 8-OHD (100 μM) treatment for 48 h. Bioinformatics analysis of DEGs showed that hemopoiesis, cell cycle regulation, nuclear factor-κB (NF-κB), and mitogen-activated protein kinase (MAPK) and Janus kinase/signal transducers and activators of transcription (JAK-STAT)-mediated apoptosis/anti-apoptosis networks were significantly regulated by 8-OHD. Western blot analysis confirmed that 8-OHD significantly induced the activation of MAPK and NF-κB signaling pathways, both of which may be responsible, at least in part, for the stimulation of apoptosis, autophagy, and differentiation in K562 cells. This is the first report on the anti-CML effects of 8-OHD and the combination of experimental and in silico analyses could provide a better understanding for the development of 8-OHD on CML therapy.
Collapse
Affiliation(s)
- Pei-Shan Wu
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan;
| | - Jui-Hung Yen
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 970, Taiwan; (J.-H.Y.); (P.-Y.C.)
- Institute of Medical Sciences, Tzu Chi University, Hualien 970, Taiwan
| | - Chih-Yang Wang
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, Taipei Medical University, Taipei 11031, Taiwan;
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei 11031, Taiwan
| | - Pei-Yi Chen
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 970, Taiwan; (J.-H.Y.); (P.-Y.C.)
- Center of Medical Genetics, Buddhist Tzu Chi General Hospital, Hualien 970, Taiwan
| | - Jui-Hsiang Hung
- Department of Biotechnology, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan;
| | - Ming-Jiuan Wu
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan;
| |
Collapse
|
15
|
Bai B, Shan L, Wang J, Hu J, Zheng W, Lv Y, Chen K, Xu D, Zhu H. Small molecule 2,3-DCPE induces S phase arrest by activating the ATM/ATR-Chk1-Cdc25A signaling pathway in DLD-1 colon cancer cells. Oncol Lett 2020; 20:294. [PMID: 33101488 PMCID: PMC7576987 DOI: 10.3892/ol.2020.12157] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 09/09/2020] [Indexed: 01/05/2023] Open
Abstract
In our previous study, it was reported that 2[[3-(2,3-dichlorophenoxy)propyl]amino]ethanol (2,3-DCPE) induces apoptosis and cell cycle arrest. The current study aimed to investigate the molecular mechanism involved in 2,3-DCPE-induced S phase arrest. The results demonstrated that 2,3-DCPE upregulated phosphorylated (p-)H2A histone family member X, a biomarker of DNA damage, in the DLD-1 colon cancer cell line. Western blotting revealed that 2,3-DCPE increased the checkpoint kinase (Chk)1 (Ser317 and Ser345) level and decreased the expression of M-phase inducer phosphatase 1 (Cdc25A) in a time-dependent manner. Subsequently, the results demonstrated that the ataxia-telangiectasia mutated (ATM) and ataxia-telangiectasia and Rad3-related (ATR) inhibitors wortmannin and caffeine had no effect on the cell cycle; however, the inhibitors partially abrogated 2,3-DCPE-induced S phase arrest. Flow cytometry assays revealed that caffeine (2 mM) reduced the proportion of S phase cells from 83 to 39.6% and that wortmannin (500 nM) reduced the proportion of S phase cells from 83 to 48.2%. Furthermore, wortmannin and caffeine inhibited the 2,3-DCPE-mediated phosphorylation of Chk1 and the degradation of Cdc25A. However, these ATM/ATR inhibitors had limited effect on 2,3-DCPE-induced apoptosis. Taken together, the data of the current study indicated that 2,3-DCPE caused DNA damage in colon cancer cells and that 2,3-DCPE-induced S phase arrest was associated with the activation of the ATM/ATR-Chk1-Cdc25A pathway.
Collapse
Affiliation(s)
- Bingjun Bai
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China.,Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang 310016, P.R. China
| | - Lina Shan
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China.,Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang 310016, P.R. China
| | - Jianhong Wang
- Department of Colorectal Surgery, Deqing People's Hospital/Deqing Campus, Sir Run Run Shaw Hospital, Huzhou, Zhejiang 313216, P.R. China
| | - Jinhui Hu
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China.,Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang 310016, P.R. China
| | - Wenqian Zheng
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China.,Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang 310016, P.R. China
| | - Yiming Lv
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China.,Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang 310016, P.R. China
| | - Kangke Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China.,Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang 310016, P.R. China
| | - Dengyong Xu
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Hongbo Zhu
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China.,Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang 310016, P.R. China
| |
Collapse
|
16
|
Carbon nanotube filler enhances incinerated thermoplastics-induced cytotoxicity and metabolic disruption in vitro. Part Fibre Toxicol 2020; 17:40. [PMID: 32787867 PMCID: PMC7424660 DOI: 10.1186/s12989-020-00371-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/28/2020] [Indexed: 11/11/2022] Open
Abstract
Background Engineered nanomaterials are increasingly being incorporated into synthetic materials as fillers and additives. The potential pathological effects of end-of-lifecycle recycling and disposal of virgin and nano-enabled composites have not been adequately addressed, particularly following incineration. The current investigation aims to characterize the cytotoxicity of incinerated virgin thermoplastics vs. incinerated nano-enabled thermoplastic composites on two in vitro pulmonary models. Ultrafine particles released from thermally decomposed virgin polycarbonate or polyurethane, and their carbon nanotube (CNT)-enabled composites were collected and used for acute in vitro exposure to primary human small airway epithelial cell (pSAEC) and human bronchial epithelial cell (Beas-2B) models. Post-exposure, both cell lines were assessed for cytotoxicity, proliferative capacity, intracellular ROS generation, genotoxicity, and mitochondrial membrane potential. Results The treated Beas-2B cells demonstrated significant dose-dependent cellular responses, as well as parent matrix-dependent and CNT-dependent sensitivity. Cytotoxicity, enhancement in reactive oxygen species, and dissipation of ΔΨm caused by incinerated polycarbonate were significantly more potent than polyurethane analogues, and CNT filler enhanced the cellular responses compared to the incinerated parent particles. Such effects observed in Beas-2B were generally higher in magnitude compared to pSAEC at treatments examined, which was likely attributable to differences in respective lung cell types. Conclusions Whilst the effect of the treatments on the distal respiratory airway epithelia remains limited in interpretation, the current in vitro respiratory bronchial epithelia model demonstrated profound sensitivity to the test particles at depositional doses relevant for occupational cohorts.
Collapse
|
17
|
AlQathama A, Shao L, Bader A, Khondkar P, Gibbons S, M Prieto J. Differential Anti-Proliferative and Anti-Migratory Activities of Ursolic Acid, 3- O-Acetylursolic Acid and Their Combination Treatments with Quercetin on Melanoma Cells. Biomolecules 2020; 10:E894. [PMID: 32545262 PMCID: PMC7356947 DOI: 10.3390/biom10060894] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/10/2020] [Accepted: 06/10/2020] [Indexed: 01/02/2023] Open
Abstract
We evaluate how 3-acetylation modulates the in vitro activity of ursolic acid in melanoma cells alone or in combination treatments with quercetin. Anti-proliferative studies on A375 cells and adult human dermal fibroblasts included analyses on cell cycle distribution, caspase activity, phosphatidylserine translocation, cell morphology and Bax/Bcl-2 protein expression. Then, 2D and 3D migration of B16F10 cells were studied using scratch and Transwell assays, respectively. Ursolic acid and 3-O-acetylursolic acid have shown similar GI50 on A375 cells (26 µM vs. 32 µM, respectively) significantly increased both early and late apoptotic populations, activated caspases 3/7 (48-72 h), and enhanced Bax whilst attenuating Bcl-2 expression. Ursolic acid caused elevation of the sub-G1 population whilst its 3-acetyl derivative arrested cell cycle at S phase and induced strong morphological changes. Combination treatments showed that ursolic acid and quercetin act synergistically in migration assays but not against cell proliferation. In summary, 3-O-acetylursolic acid maintains the potency and overall apoptotic mechanism of the parent molecule with a more aggressive influence on the morphology of A375 melanoma cells but the 3-acetylation suppresses its anti-migratory properties. We also found that ursolic acid can act in synergy with quercetin to reduce cell migration.
Collapse
Affiliation(s)
- Aljawharah AlQathama
- School of Pharmacy, University College London, London WC1N 1AX, UK; (L.S.); (P.K.); (S.G.)
- Department of Pharmacognosy, Pharmacy College, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Luying Shao
- School of Pharmacy, University College London, London WC1N 1AX, UK; (L.S.); (P.K.); (S.G.)
| | - Ammar Bader
- Department of Pharmacognosy, Pharmacy College, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Proma Khondkar
- School of Pharmacy, University College London, London WC1N 1AX, UK; (L.S.); (P.K.); (S.G.)
| | - Simon Gibbons
- School of Pharmacy, University College London, London WC1N 1AX, UK; (L.S.); (P.K.); (S.G.)
- School of Pharmacy, University East Anglia, Norwich NR4 7TJ, UK
| | - Jose M Prieto
- School of Pharmacy, University College London, London WC1N 1AX, UK; (L.S.); (P.K.); (S.G.)
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool L3 3AF, UK
| |
Collapse
|
18
|
Bordoloi D, Banik K, Padmavathi G, Vikkurthi R, Harsha C, Roy NK, Singh AK, Monisha J, Wang H, Kumar AP, Kunnumakkara AB. TIPE2 Induced the Proliferation, Survival, and Migration of Lung Cancer Cells Through Modulation of Akt/mTOR/NF-κB Signaling Cascade. Biomolecules 2019; 9:E836. [PMID: 31817720 PMCID: PMC6995575 DOI: 10.3390/biom9120836] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/27/2019] [Accepted: 12/02/2019] [Indexed: 12/17/2022] Open
Abstract
Lung cancer represents the most common cause of cancer deaths in the world, constituting around 11.6% of all new cancer cases and 18.4% of cancer-related deaths. The propensity for early spread, lack of suitable biomarkers for early diagnosis, as well as prognosis and ineffective existing therapies, contribute to the poor survival rate of lung cancer. Therefore, there is an urgent need to develop novel biomarkers for early diagnosis and prognosis which in turn can facilitate newer therapeutic avenues for the management of this aggressive neoplasm. TIPE2 (tumor necrosis factor-α-induced protein 8-like 2), a recently identified cytoplasmic protein, possesses enormous potential in this regard. Immunohistochemical analysis showed that TIPE2 was significantly upregulated in different stages and grades of lung cancer tissues compared to normal lung tissues, implying its involvement in the positive regulation of lung cancer. Further, knockout of TIPE2 resulted in significantly reduced proliferation, survival, and migration of human lung cancer cells through modulation of the Akt/mTOR/NF-κB signaling axis. In addition, knockout of TIPE2 also caused arrest in the S phase of the cell cycle of lung cancer cells. As tobacco is the most predominant risk factor for lung cancer, we therefore evaluated the effect of TIPE2 in tobacco-mediated lung carcinogenesis as well. Our results showed that TIPE2 was involved in nicotine-, nicotine-derived nitrosamine ketone (NNK)-, N-nitrosonornicotine (NNN)-, and benzo[a]pyrene (BaP)-mediated lung cancer through inhibited proliferation, survival, and migration via modulation of nuclear factor kappa B (NF-κB)- and NF-κB-regulated gene products, which are involved in the regulation of diverse processes in lung cancer cells. Taken together, TIPE2 possesses an important role in the development and progression of lung cancer, particularly in tobacco-promoted lung cancer, and hence, specific targeting of it holds an enormous prospect in newer therapeutic interventions in lung cancer. However, these findings need to be validated in the in vivo and clinical settings to fully establish the diagnostic and prognostic importance of TIPE2 against lung cancer.
Collapse
Affiliation(s)
- Devivasha Bordoloi
- Cancer Biology Laboratory and DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India; (D.B.); (K.B.); (G.P.); (R.V.); (C.H.); (N.K.R.); (A.K.S.); (J.M.)
| | - Kishore Banik
- Cancer Biology Laboratory and DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India; (D.B.); (K.B.); (G.P.); (R.V.); (C.H.); (N.K.R.); (A.K.S.); (J.M.)
| | - Ganesan Padmavathi
- Cancer Biology Laboratory and DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India; (D.B.); (K.B.); (G.P.); (R.V.); (C.H.); (N.K.R.); (A.K.S.); (J.M.)
| | - Rajesh Vikkurthi
- Cancer Biology Laboratory and DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India; (D.B.); (K.B.); (G.P.); (R.V.); (C.H.); (N.K.R.); (A.K.S.); (J.M.)
| | - Choudhary Harsha
- Cancer Biology Laboratory and DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India; (D.B.); (K.B.); (G.P.); (R.V.); (C.H.); (N.K.R.); (A.K.S.); (J.M.)
| | - Nand Kishor Roy
- Cancer Biology Laboratory and DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India; (D.B.); (K.B.); (G.P.); (R.V.); (C.H.); (N.K.R.); (A.K.S.); (J.M.)
| | - Anuj Kumar Singh
- Cancer Biology Laboratory and DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India; (D.B.); (K.B.); (G.P.); (R.V.); (C.H.); (N.K.R.); (A.K.S.); (J.M.)
| | - Javadi Monisha
- Cancer Biology Laboratory and DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India; (D.B.); (K.B.); (G.P.); (R.V.); (C.H.); (N.K.R.); (A.K.S.); (J.M.)
| | - Hong Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore;
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore 138602, Singapore
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore;
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory and DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India; (D.B.); (K.B.); (G.P.); (R.V.); (C.H.); (N.K.R.); (A.K.S.); (J.M.)
| |
Collapse
|
19
|
Higginsianins A and B, two fungal diterpenoid α-pyrones with cytotoxic activity against human cancer cells. Toxicol In Vitro 2019; 61:104614. [DOI: 10.1016/j.tiv.2019.104614] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/12/2019] [Accepted: 07/28/2019] [Indexed: 01/12/2023]
|
20
|
Volkova YA, Kozlov AS, Kolokolova MK, Uvarov DY, Gorbatov SA, Andreeva OE, Scherbakov AM, Zavarzin IV. Steroidal N-Sulfonylimidates: Synthesis and biological evaluation in breast cancer cells. Eur J Med Chem 2019; 179:694-706. [DOI: 10.1016/j.ejmech.2019.06.048] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/10/2019] [Accepted: 06/17/2019] [Indexed: 02/08/2023]
|
21
|
The synergistic effect of PFK15 with metformin exerts anti-myeloma activity via PFKFB3. Biochem Biophys Res Commun 2019; 515:332-338. [DOI: 10.1016/j.bbrc.2019.05.136] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 05/21/2019] [Indexed: 02/08/2023]
|
22
|
Ryu H, Jin H, Ho JN, Bae J, Lee E, Lee SE, Lee S. Suberoylanilide Hydroxamic Acid Can Re-sensitize a Cisplatin-Resistant Human Bladder Cancer. Biol Pharm Bull 2019; 42:66-72. [PMID: 30606990 DOI: 10.1248/bpb.b18-00545] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cisplatin chemotherapy is the standard treatment for metastatic urothelial carcinoma. Although there are second-line chemotherapeutic agents approved by the U.S. Food and Drug Administration (FDA) such as those targeting programmed death-ligand 1 (PD-L1), more effective pharmacotherapy is required for cisplatin-resistant bladder cancer due to its limited overall survival and progression-free survival. The synergistic anti-cancer effect of cisplatin and suberoylanilide hydroxamic acid (SAHA) in cisplatin-resistant bladder cancer cells (T24R2) was examined. Tumor cell proliferation and cell cycle was examined using the cell counting kit (CCK)-8 assays and flow cytometry, respectively. Synergism was examined using the combination index (CI). CCK-8 assay and CI test were used to observe the strong synergistic anti-cancer effect between SAHA and cisplatin. Activation of caspase mediated apoptosis, down-regulated expression of the anti-apoptotic B-cell lymphoma-2 (Bcl-2) and up-regulated expression of pro-apoptotic Bcl-2-associated death promoter (BAD) were observed in Western blot. SAHA synergistically could partially re-sensitize cisplatin-resistant bladder cancer cells (T24R2) through the cell cycle arrest and induction of apoptosis pathway. SAHA-based treatment could be a potential treatment regimen in patients with cisplatin resistant bladder cancer.
Collapse
Affiliation(s)
- Hoyoung Ryu
- Department of Urology, Seoul National University Bundang Hospital
| | - Hyunjin Jin
- Department of Urology, Seoul National University Bundang Hospital
| | - Jin-Nyoung Ho
- Department of Urology, Seoul National University Bundang Hospital
| | - Jungbum Bae
- Department of Urology, Seoul National University Bundang Hospital
| | - Eunsik Lee
- Department of Urology, Seoul National University Bundang Hospital
| | - Sang Eun Lee
- Department of Urology, Seoul National University Bundang Hospital
| | - Sangchul Lee
- Department of Urology, Seoul National University Bundang Hospital
| |
Collapse
|
23
|
Takami M, Cunha C, Motohashi S, Nakayama T, Iwashima M. TGF-β suppresses RasGRP1 expression and supports regulatory T cell resistance against p53-induced CD28-dependent T-cell apoptosis. Eur J Immunol 2018; 48:1938-1943. [PMID: 30298904 PMCID: PMC6368088 DOI: 10.1002/eji.201847587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 08/29/2018] [Accepted: 10/05/2018] [Indexed: 11/06/2022]
Abstract
Thymus-derived regulatory T cells (tTregs) play pivotal roles in immunological self-tolerance and homeostasis. A majority of tTregs are reactive to self-antigens and are constantly exposed to antigenic stimulation. Despite this continuous stimulation, tTreg and conventional T-cell populations remain balanced during homeostasis, but the mechanisms controlling this balance are unknown. We previously reported a form of activation-induced cell death, which is dependent on p53 (p53-induced CD28-dependent T-cell apoptosis, PICA). Under PICA-inducing conditions, tTregs survive while a majority of conventional T cells undergo apoptosis, suggesting there is a survival mechanism that protects tTregs. Here, we report that the expression of RasGRP1 (Ras guanyl-releasing protein 1) is required for PICA, as conventional T cells isolated from RasGRP1-deficient mice become resistant to PICA. After continuous stimulation, tTregs express a substantially lower amount of RasGRP1 compared to conventional T cells. This reduced expression of RasGRP1 is dependent on TGF-β, as addition of TGF-β to conventional T cells reduces RasGRP1 expression. Conversely, RasGRP1 expression in tTregs increases when TGF-β signaling is inhibited. Together, these data show that RasGRP1 expression is repressed in tTregs by TGF-β signaling and suggests that reduced RasGRP1 expression is critical for tTregs to resist apoptosis caused by continuous antigen exposure.
Collapse
Affiliation(s)
- Mariko Takami
- Department of Microbiology and Immunology, Loyola University, Chicago, IL, USA
- Van Kampen Cardio Pulmonary Research Laboratory, Loyola University, Chicago, IL, USA
- Department of Medical Immunology, Chiba University, Japan
| | - Christina Cunha
- Department of Microbiology and Immunology, Loyola University, Chicago, IL, USA
| | | | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Japan
| | - Makio Iwashima
- Department of Microbiology and Immunology, Loyola University, Chicago, IL, USA
- Van Kampen Cardio Pulmonary Research Laboratory, Loyola University, Chicago, IL, USA
| |
Collapse
|
24
|
NSC 95397 Suppresses Proliferation and Induces Apoptosis in Colon Cancer Cells through MKP-1 and the ERK1/2 Pathway. Int J Mol Sci 2018; 19:ijms19061625. [PMID: 29857489 PMCID: PMC6032145 DOI: 10.3390/ijms19061625] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/23/2018] [Accepted: 05/24/2018] [Indexed: 01/21/2023] Open
Abstract
NSC 95397, a quinone-based small molecule compound, has been identified as an inhibitor for dual-specificity phosphatases, including mitogen-activated protein kinase phosphatase-1 (MKP-1). MKP-1 is known to inactivate mitogen-activated protein kinases by dephosphorylating both of their threonine and tyrosine residues. Moreover, owing to their participation in tumorigenesis and drug resistance in colon cancer cells, MKP-1 is an attractive therapeutic target for colon cancer treatment. We therefore investigated the inhibitory activity of NSC 95397 against three colon cancer cell lines including SW480, SW620, and DLD-1, and their underlying mechanisms. The results demonstrated that NSC 95397 reduced cell viability and anchorage-independent growth of all the three colon cancer cell lines through inhibited proliferation and induced apoptosis via regulating cell-cycle-related proteins, including p21, cyclin-dependent kinases, and caspases. Besides, by using mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) inhibitor U0126, we provided mechanistic evidence that the antineoplastic effects of NSC 95397 were achieved via inhibiting MKP-1 activity followed by ERK1/2 phosphorylation. Conclusively, our results indicated that NSC 95397 might serve as an effective therapeutic intervention for colon cancer through regulating MKP-1 and ERK1/2 pathway.
Collapse
|
25
|
Kim H, Yi SS, Lee HK, Heo TH, Park SK, Jun HS, Song KD, Kim SJ. Antiproliferative Effect of Vine Stem Extract from Spatholobus Suberectus Dunn on Rat C6 Glioma Cells Through Regulation of ROS, Mitochondrial Depolarization, and P21 Protein Expression. Nutr Cancer 2018; 70:605-619. [PMID: 29668336 DOI: 10.1080/01635581.2018.1460673] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The vine stem of Spatholobus suberectus Dunn (SS) is used as a traditional herbal medicine in China. Chinese herbal medicines are well known as natural bioactive compounds that can be used as new medicines, and their antioxidant and anticancer effects have also been reported. This study aimed to examine the anticancer effect of a high-pressure hot-water SS extract on rat C6 glioma cells. The SS extract effectively suppressed the viability and proliferation of C6 glioma cells through an antioxidant effect. Reactive oxygen species (ROS) levels in cancer cells are higher than that in normal cells. If the ROS level falls below that required for the growth of cancer cells, their rapid proliferation and growth can be suppressed. We also measured the induction of mitochondrial membrane depolarization and cell cycle arrest effect caused by the SS extract in C6 glioma cells through a FACS analysis. In addition, we observed an increase in STAT3, p53, E2F1, and p21 mRNA expression and a decrease in Bcl-2 mRNA expression by quantitative PCR. An increase in p21 protein expression of over 83% was observed through western blot analysis. All these data support the fact that the high-pressure hot-water SS extract has the potential to be used for glioma treatment.
Collapse
Affiliation(s)
- Hyungkuen Kim
- a Department of Biotechnology , Hoseo University , Asan , Chungnam , Republic of Korea
| | - Sun Shin Yi
- b Department of Biomedical Laboratory Science , College of Biomedical Sciences, Soonchunhyang University , Asan , Chungnam , Republic of Korea
| | - Hak-Kyo Lee
- c Department of Animal Biotechnology , Chonbuk National University , Jeonju , Jeonbuk , Republic of Korea
| | - Tae-Hwe Heo
- d Lab of Immunology, Integrated Research Institute of Pharmaceutical Sciences, College of Pharmacy, The Catholic University of Korea , Bucheon , Gyeonggi , Republic of Korea
| | - Sang-Kyu Park
- e Department of Medical Biotechnology , College of Medical Sciences, Soonchunhyang University , Asan , Chungnam , Republic of Korea
| | - Hyun Sik Jun
- f Department of Biotechnology and Bioinformatics , College of Science and Technology, Korea University , Sejong , Republic of Korea
| | - Ki Duk Song
- c Department of Animal Biotechnology , Chonbuk National University , Jeonju , Jeonbuk , Republic of Korea
| | - Sung-Jo Kim
- a Department of Biotechnology , Hoseo University , Asan , Chungnam , Republic of Korea
| |
Collapse
|
26
|
Gardella KA, Muro I, Fang G, Sarkar K, Mendez O, Wright CW. Aryl hydrocarbon receptor nuclear translocator (ARNT) isoforms control lymphoid cancer cell proliferation through differentially regulating tumor suppressor p53 activity. Oncotarget 2017; 7:10710-22. [PMID: 26909609 PMCID: PMC4905433 DOI: 10.18632/oncotarget.7539] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 01/22/2016] [Indexed: 12/25/2022] Open
Abstract
The aryl hydrocarbon receptor nuclear translocator (ARNT) is involved in xenobiotic and hypoxic responses, and we previously showed that ARNT also regulates nuclear factor-κB (NF-κB) signaling by altering the DNA binding activity of the RelB subunit. However, our initial study of ARNT-mediated RelB modulation was based on simultaneous suppression of the two ARNT isoforms, isoform 1 and 3, and precluded the examination of their individual functions. We find here that while normal lymphocytes harbor equal levels of isoform 1 and 3, lymphoid malignancies exhibit a shift to higher levels of ARNT isoform 1. These elevated levels of ARNT isoform 1 are critical to the proliferation of these cancerous cells, as suppression of isoform 1 in a human multiple myeloma (MM) cell line, and an anaplastic large cell lymphoma (ALCL) cell line, triggered S-phase cell cycle arrest, spontaneous apoptosis, and sensitized cells to doxorubicin treatment. Furthermore, co-suppression of RelB or p53 with ARNT isoform 1 prevented cell cycle arrest and blocked doxorubicin induced apoptosis. Together our findings reveal that certain blood cancers rely on ARNT isoform 1 to potentiate proliferation by antagonizing RelB and p53-dependent cell cycle arrest and apoptosis. Significantly, our results identify ARNT isoform 1 as a potential target for anticancer therapies.
Collapse
Affiliation(s)
- Kacie A Gardella
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, USA
| | - Israel Muro
- Division of Pharmacology and Toxicology, and The Center for Molecular and Cellular Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| | - Gloria Fang
- Division of Pharmacology and Toxicology, and The Center for Molecular and Cellular Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| | - Krishnakali Sarkar
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, USA
| | - Omayra Mendez
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, USA
| | - Casey W Wright
- Division of Pharmacology and Toxicology, and The Center for Molecular and Cellular Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA.,Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
27
|
Huang D, Feng X, Liu Y, Deng Y, Chen H, Chen D, Fang L, Cai Y, Liu H, Wang L, Wang J, Yang Z. AQP9-induced cell cycle arrest is associated with RAS activation and improves chemotherapy treatment efficacy in colorectal cancer. Cell Death Dis 2017. [PMID: 28640255 PMCID: PMC5520935 DOI: 10.1038/cddis.2017.282] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aquaporin-9 (AQP9) expression is associated with arsenic sensitivity in leukemia cells. However, the role of AQP9 in regulating tumor sensitivity to adjuvant chemotherapy in colorectal cancer (CRC) has not been elucidated. In this study, we demonstrated that AQP9 can serve as an independent predictive marker for adjuvant chemotherapy in CRC. Patients with high AQP9 expression had higher rate of disease-free survival (DFS) than those with low AQP9 expression. Upregulation of AQP9 was associated with enhanced chemosensitivity to 5-fluorouracil (5-FU) both in vitro and in vivo. Overexpression of AQP9 resulted in an increased intracellular level of 5-FU in CRC cells, hence leading to a higher percentage of apoptosis after 5-FU treatment. Moreover, AQP9 is positively associated with RAS activation and other downstream signaling molecules in CRC. AQP9 overexpression resulted in p21 upregulation and induced S-phase arrest. Taken together, AQP9 enhances the cytotoxic response to 5-FU in CRC cells by simultaneously inducing S-phase arrest via activation of RAS signaling and facilitating drug uptake. Our results suggest that AQP9 might be a novel predictor for the benefit of 5-FU-based chemotherapy in CRC. The identification of AQP9-induced tumor sensitivity to 5-FU highlights the role of AQP9 in regulating chemosensitivity in CRC.
Collapse
Affiliation(s)
- Dandan Huang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| | - Xingzhi Feng
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| | - Yiting Liu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| | - Yanhong Deng
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China.,Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Hao Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| | - Daici Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| | - Lekun Fang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| | - Yue Cai
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China.,Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Huanliang Liu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| | - Lei Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| | - Jianping Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| | - Zihuan Yang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| |
Collapse
|
28
|
Wang Y, Compton C, Rankin GO, Cutler SJ, Rojanasakul Y, Tu Y, Chen YC. 3-Hydroxyterphenyllin, a natural fungal metabolite, induces apoptosis and S phase arrest in human ovarian carcinoma cells. Int J Oncol 2017; 50:1392-1402. [PMID: 28259974 PMCID: PMC5363874 DOI: 10.3892/ijo.2017.3894] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 02/13/2017] [Indexed: 01/20/2023] Open
Abstract
In the present study, we evaluated 3-Hydroxyter-phenyllin (3-HT) as a potential anticancer agent using the human ovarian cancer cells A2780/CP70 and OVCAR-3, and normal human epithelial ovarian cells IOSE-364 as an in vitro model. 3-HT suppressed proliferation and caused cytotoxicity against A2780/CP70 and OVCAR-3 cells, while it exhibited lower cytotoxicity in IOSE-364 cells. Subsequently, we found that 3-HT induced S phase arrest and apoptosis in a dose-independent manner. Further investigation revealed that S phase arrest was related with DNA damage which mediated the ATM/p53/Chk2 pathway. Downregulation of cyclin D1, cyclin A2, cyclin E1, CDK2, CDK4 and Cdc25C, and the upregulation of Cdc25A and cyclin B1 led to the accumulation of cells in S phase. The apoptotic effect was confirmed by Hoechst 33342 staining, depolarization of mitochondrial membrane potential and activation of cleaved caspase-3 and PARP1. Additional results revealed both intrinsic and extrinsic apoptotic pathways were involved. The intrinsic apoptotic pathway was activated through decreasing the protein levels of Bcl2, Bcl-xL and procaspase-9 and increasing the protein level of Puma. The induction of DR5 and DR4 indicated that the extrinsic apoptotic pathway was also activated. Induction of ROS and activation of ERK were observed in ovarian cancer cells. We therefore concluded that 3-HT possessed anti-proliferative effect on A2780/CP70 and OVCAR-3 cells, induced S phase arrest and caused apoptosis. Taken together, we propose that 3-HT shows promise as a therapeutic candidate for treating ovarian cancer.
Collapse
Affiliation(s)
- Yaomin Wang
- Department of Tea Science, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Casey Compton
- College of Science, Technology and Mathematics, Alderson Broaddus University, Philippi, WV 26416, USA
| | - Gary O Rankin
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Stephen J Cutler
- Department of BioMolecular Sciences, University of Mississippi, University, MS 38677, USA
| | - Yon Rojanasakul
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV 26506, USA
| | - Youying Tu
- Department of Tea Science, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Yi Charlie Chen
- College of Science, Technology and Mathematics, Alderson Broaddus University, Philippi, WV 26416, USA
| |
Collapse
|
29
|
Ding Y, Gu Z, Wang Y, Wang S, Chen H, Zhang H, Chen W, Chen YQ. Clove extract functions as a natural fatty acid synthesis inhibitor and prevents obesity in a mouse model. Food Funct 2017; 8:2847-2856. [DOI: 10.1039/c7fo00096k] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Numerous medicinal plants have been reported to prevent various chronic diseases.
Collapse
Affiliation(s)
- Yiran Ding
- State Key Laboratory of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
- School of Food Science and Technology
| | - Zhennan Gu
- State Key Laboratory of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
- School of Food Science and Technology
| | - Yihe Wang
- State Key Laboratory of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
- School of Food Science and Technology
| | - Shunhe Wang
- State Key Laboratory of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
- School of Food Science and Technology
| | - Haiqin Chen
- State Key Laboratory of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
- School of Food Science and Technology
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
- School of Food Science and Technology
| | - Wei Chen
- State Key Laboratory of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
- School of Food Science and Technology
| | - Yong Q. Chen
- State Key Laboratory of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
- School of Food Science and Technology
| |
Collapse
|
30
|
Reza AMMT, Choi YJ, Yasuda H, Kim JH. Human adipose mesenchymal stem cell-derived exosomal-miRNAs are critical factors for inducing anti-proliferation signalling to A2780 and SKOV-3 ovarian cancer cells. Sci Rep 2016; 6:38498. [PMID: 27929108 PMCID: PMC5143979 DOI: 10.1038/srep38498] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 11/11/2016] [Indexed: 02/07/2023] Open
Abstract
An enigmatic question exists concerning the pro- or anti-cancer status of mesenchymal stem cells (MSCs). Despite growing interest, this question remains unanswered, and the debate became intensified with new evidences backing each side. Here, we showed that human adipose MSC (hAMSC)-derived conditioned medium (CM) exhibited inhibitory effects on A2780 human ovarian cancer cells by blocking the cell cycle, and activating mitochondria-mediated apoptosis signalling. Explicitly, we demonstrated that exosomes, an important biological component of hAMSC-CM, could restrain proliferation, wound-repair and colony formation ability of A2780 and SKOV-3 cancer cells. Furthermore, hAMSC-CM-derived exosomes induced apoptosis signalling by upregulating different pro-apoptotic signalling molecules, such as BAX, CASP9, and CASP3, as well as downregulating the anti-apoptotic protein BCL2. More specifically, cancer cells exhibited reduced viability following fresh or protease-digested exosome treatment; however, treatment with RNase-digested exosomes could not inhibit the proliferation of cancer cells. Additionally, sequencing of exosomal RNAs revealed a rich population of microRNAs (miRNAs), which exhibit anti-cancer activities by targeting different molecules associated with cancer survival. Our findings indicated that exosomal miRNAs are important players involved in the inhibitory influence of hAMSC-CM towards ovarian cancer cells. Therefore, we believe that these comprehensive results will provide advances concerning ovarian cancer research and treatment.
Collapse
Affiliation(s)
- Abu Musa Md Talimur Reza
- Department of Stem Cell and Regenerative Biology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul 143-701, Republic of Korea
| | - Yun-Jung Choi
- Department of Stem Cell and Regenerative Biology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul 143-701, Republic of Korea
| | - Hideyo Yasuda
- Department of Stem Cell and Regenerative Biology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul 143-701, Republic of Korea
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul 143-701, Republic of Korea
| |
Collapse
|
31
|
Yang Z, Zhao Y, Yao Y, Li J, Wang W, Wu X. Equol Induces Mitochondria-Dependent Apoptosis in Human Gastric Cancer Cells via the Sustained Activation of ERK1/2 Pathway. Mol Cells 2016; 39:742-749. [PMID: 27802587 PMCID: PMC5104882 DOI: 10.14348/molcells.2016.0162] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 09/01/2016] [Accepted: 09/22/2016] [Indexed: 11/27/2022] Open
Abstract
The cancer chemo-preventive effects of equol have been demonstrated for a wide variety of experimental tumours. In a previous study, we found that equol inhibited proliferation and induced apoptotic death of human gastric cancer MGC-803 cells. However, the mechanisms underlying equol-mediated apoptosis have not been well understood. In the present study, the dual AO (acridine orange)/EB (ethidium bromide) fluorescent assay, the comet assay, MTS, western blotting and flow cytometric assays were performed to further investigate the pro-apoptotic effect of equol and its associated mechanisms in MGC-803 cells. The results demonstrated that equol induced an apoptotic nuclear morphology revealed by AO/EB staining, the presence of a comet tail, the cleavage of caspase-3 and PARP and the depletion of cIAP1, indicating its pro-apoptotic effect. In addition, equol-induced apoptosis involves the mitochondria-dependent cell-death pathway, evidenced by the depolarization of the mitochondrial membrane potential, the cleavage of caspase-9 and the depletion of Bcl-xL and full-length Bid. Moreover, treating MGC-803 cells with equol induced the sustained activation of extracellular signal-regulated kinase (ERK), and inhibiting ERK by U0126, a MEK/ERK pathway inhibitor, significantly attenuated the equol-induced cell apoptosis. These results suggest that equol induces mitochondria-dependent apoptosis in human gastric cancer MGC-803 cells via the sustained activation of the ERK1/2 pathway. Therefore, equol may be a novel candidate for the chemoprevention and therapy of gastric cancer.
Collapse
Affiliation(s)
- Zhiping Yang
- The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003
P.R. China
| | - Yan Zhao
- The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003
P.R. China
| | - Yahong Yao
- The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003
P.R. China
| | - Jun Li
- The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003
P.R. China
| | - Wangshi Wang
- The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003
P.R. China
| | - Xiaonan Wu
- Xiamen Medical College, Xiamen, Fujian 350108,
P.R. China
| |
Collapse
|
32
|
Sharma V, Kumar L, Mohanty SK, Maikhuri JP, Rajender S, Gupta G. Sensitization of androgen refractory prostate cancer cells to anti-androgens through re-expression of epigenetically repressed androgen receptor - Synergistic action of quercetin and curcumin. Mol Cell Endocrinol 2016; 431:12-23. [PMID: 27132804 DOI: 10.1016/j.mce.2016.04.024] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 04/02/2016] [Accepted: 04/27/2016] [Indexed: 01/06/2023]
Abstract
Epigenetic repression of Androgen Receptor (AR) gene by hypermethylation of its promoter causes resistance in prostate cancer (CaP) to androgen deprivation therapy with anti-androgens. Some dietary phytocompounds like quercetin (Q) and curcumin (C) with reported DNMT-inhibitory activity were tested for their ability to re-express the AR in AR-negative CaP cell lines PC3 and DU145. Combined treatment with Q+C was much more effective than either Q or C in inhibiting DNMT, causing global hypomethylation, restoring AR mRNA and protein levels and causing apoptosis via mitochondrial depolarization of PC3 and DU145. The functional AR protein expressed in Q+C treated cells sensitized them to dihydrotestosterone (DHT)-induced proliferation, bicalutamide-induced apoptosis, bound to androgen response element to increase luciferase activity in gene reporter assay and was susceptible to downregulation by AR siRNA. Bisulfite sequencing revealed high methylation of AR promoter CpG sites in AR-negative DU145 and PC3 cell lines that was significantly demethylated by Q+C treatment, which restored AR expression. Notable synergistic effects of Q+C combination in re-sensitizing androgen refractory CaP cells to AR-mediated apoptosis, their known safety in clinical use, and epidemiological evidences relating their dietary consumption with lower cancer incidences indicate their potential for use in chemoprevention of androgen resistance in prostate cancer.
Collapse
Affiliation(s)
- Vikas Sharma
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Lokesh Kumar
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Sujit K Mohanty
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Jagdamba P Maikhuri
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Singh Rajender
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Gopal Gupta
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India.
| |
Collapse
|
33
|
Wani ZA, Guru SK, Rao AVS, Sharma S, Mahajan G, Behl A, Kumar A, Sharma PR, Kamal A, Bhushan S, Mondhe DM. A novel quinazolinone chalcone derivative induces mitochondrial dependent apoptosis and inhibits PI3K/Akt/mTOR signaling pathway in human colon cancer HCT-116 cells. Food Chem Toxicol 2015; 87:1-11. [PMID: 26615871 DOI: 10.1016/j.fct.2015.11.016] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Revised: 11/09/2015] [Accepted: 11/19/2015] [Indexed: 11/17/2022]
Abstract
We have synthesized a novel quinazolinone chalcone derivative (QC) and first time reported its in-vitro and in-vivo anticancer potential. It inhibited the cell proliferation of different cancer cell lines like PC-3, Panc-1, Mia-Paca-2, A549, MCF-7 and HCT-116. It induces apoptosis as measured by several biological endpoints such as apoptotic body formation, evident by Hoechst and scanning electron microscopy, enhanced annexinV-FITC binding of the cells, increased sub-G0 cell fraction, loss of mitochondrial membrane potential (Δψm), reduction of Bcl-2/Bax ratio, activation of caspase-9, caspase-3 and PARP-1 (poly-ADP Ribose polymerase) cleavage in HCT-116 cells. In spite of apoptosis, QC significantly hammers the downstream and upstream signaling cascade of PI3K/Akt/mTOR pathway and cell cycle regulator Skp-2, p21 and p27. Interestingly, QC induces the S and G2/M phase of HCT-116 cells at experimental doses. QC inhibits the tumor growth of Ehrlich ascites carcinoma (EAC), Ehrlich tumor (ET, solid) and sarcoma-180(solid) mice models. Furthermore, it was found to be non-toxic as no animal mortality (0/7) occurred during experimental doses. The present study provides an insight of anticancer potential of QC, which may be useful in managing and treating cancer.
Collapse
Affiliation(s)
- Zahoor Ahmad Wani
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
| | - Santosh Kumar Guru
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
| | - A V Subba Rao
- Indian Institute of Chemical Technology, Tarnaka, Hyderabad, 500607, India
| | - Sonia Sharma
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
| | - Girish Mahajan
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
| | - Akanksha Behl
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
| | - Ashok Kumar
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
| | - P R Sharma
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
| | - Ahmed Kamal
- Indian Institute of Chemical Technology, Tarnaka, Hyderabad, 500607, India
| | - Shashi Bhushan
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India.
| | - Dilip M Mondhe
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India.
| |
Collapse
|
34
|
Cruz e Carvalho A, Márquez CAP, Azevedo RB, Joanitti GA, Pires Júnior OR, Fontes W, Castro MS. Cytotoxic Activity and Antiproliferative Effects of Crude Skin Secretion from Physalaemus nattereri (Anura: Leptodactylidae) on in vitro Melanoma Cells. Toxins (Basel) 2015; 7:3989-4005. [PMID: 26457717 PMCID: PMC4626716 DOI: 10.3390/toxins7103989] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/24/2015] [Accepted: 08/27/2015] [Indexed: 11/23/2022] Open
Abstract
Anuran secretions are rich sources of bioactive molecules, including antimicrobial and antitumoral compounds. The aims of this study were to investigate the therapeutic potential of Physalaemus nattereri skin secretion against skin cancer cells, and to assess its cytotoxic action mechanisms on the murine melanoma cell line B16F10. Our results demonstrated that the crude secretion reduced the viability of B16F10 cells, causing changes in cell morphology (e.g., round shape and structure shrinkage), reduction in mitochondrial membrane potential, increase in phosphatidylserine exposure, and cell cycle arrest in S-phase. Together, these changes suggest that tumor cells die by apoptosis. This skin secretion was also subjected to chromatographic fractioning using RP-HPLC, and eluted fractions were assayed for antiproliferative and antibacterial activities. Three active fractions showed molecular mass components in a range compatible with peptides. Although the specific mechanisms causing the reduced cell viability and cytotoxicity after the treatment with crude secretion are still unknown, it may be considered that molecules, such as the peptides found in the secretion, are effective against B16F10 tumor cells. Considering the growing need for new anticancer drugs, data presented in this study strongly reinforce the validity of P. nattereri crude secretion as a rich source of new anticancer molecules.
Collapse
Affiliation(s)
- Andréa Cruz e Carvalho
- Laboratory of Toxinology, Department of Physiological Sciences/IB, University of Brasília, Brasília/DF, CEP 70.910-900, Brazil.
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology/IB, University of Brasília, Brasília/DF, CEP 70.910-900, Brazil.
| | - César Augusto Prías Márquez
- Laboratory of Toxinology, Department of Physiological Sciences/IB, University of Brasília, Brasília/DF, CEP 70.910-900, Brazil.
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology/IB, University of Brasília, Brasília/DF, CEP 70.910-900, Brazil.
| | - Ricardo Bentes Azevedo
- Department of Genetics and Morphology/IB, University of Brasília, Brasília/DF, CEP 70.910-900, Brazil.
| | - Graziella Anselmo Joanitti
- Department of Genetics and Morphology/IB, University of Brasília, Brasília/DF, CEP 70.910-900, Brazil.
- Faculty of Ceilândia, University of Brasília, Ceilândia/DF, CEP 72.220-140, Brazil.
| | - Osmindo Rodrigues Pires Júnior
- Laboratory of Toxinology, Department of Physiological Sciences/IB, University of Brasília, Brasília/DF, CEP 70.910-900, Brazil.
| | - Wagner Fontes
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology/IB, University of Brasília, Brasília/DF, CEP 70.910-900, Brazil.
| | - Mariana S Castro
- Laboratory of Toxinology, Department of Physiological Sciences/IB, University of Brasília, Brasília/DF, CEP 70.910-900, Brazil.
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology/IB, University of Brasília, Brasília/DF, CEP 70.910-900, Brazil.
| |
Collapse
|
35
|
Kusaczuk M, Krętowski R, Bartoszewicz M, Cechowska-Pasko M. Phenylbutyrate-a pan-HDAC inhibitor-suppresses proliferation of glioblastoma LN-229 cell line. Tumour Biol 2015; 37:931-42. [PMID: 26260271 PMCID: PMC4841856 DOI: 10.1007/s13277-015-3781-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 07/08/2015] [Indexed: 12/19/2022] Open
Abstract
Phenylbutyrate (PBA) is a histone deacetylase inhibitor known for inducing differentiation, cell cycle arrest, and apoptosis in various cancer cells. However, the effects of PBA seem to be very cell-type-specific and sometimes limited exclusively to a particular cell line. Here, we provided novel information concerning cellular effects of PBA in LN-229 and LN-18 glioblastoma cell lines which have not been previously evaluated in context of PBA exposure. We found that LN-18 cells were PBA-insensitive even at high concentrations of PBA. In contrary, in LN-229 cells, 5 and 15 mmol/L PBA inhibited cell growth and proliferation mainly by causing prominent changes in cell morphology and promoting S- and G2/M-dependent cell cycle arrest. Moreover, we observed nearly a 3-fold increase in apoptosis of LN-229 cells treated with 15 mmol/L PBA, in comparison to control. Furthermore, PBA was found to up-regulate the expression of p21 whereas p53 expression level remained unchanged. We also showed that PBA down-regulated the expression of the anti-apoptotic genes Bcl-2/Bcl-XL, however without affecting the expression of pro-apoptotic Bax and Bim. Taken together, our results suggest that PBA might potentially be considered as an agent slowing-down the progress of glioblastoma; however, further analyses are still needed to comprehensively resolve the nature of its activity in this type of cancer.
Collapse
Affiliation(s)
- Magdalena Kusaczuk
- Department of Pharmaceutical Biochemistry, Medical University of Białystok, Mickiewicza 2A, 15-222, Białystok, Poland.
| | - Rafał Krętowski
- Department of Pharmaceutical Biochemistry, Medical University of Białystok, Mickiewicza 2A, 15-222, Białystok, Poland
| | - Marek Bartoszewicz
- Department of Microbiology, Institute of Biology, University of Białystok, Białystok, Poland
| | - Marzanna Cechowska-Pasko
- Department of Pharmaceutical Biochemistry, Medical University of Białystok, Mickiewicza 2A, 15-222, Białystok, Poland
| |
Collapse
|
36
|
Zhang Y, Xie RF, Xiao QG, Li R, Shen XL, Zhu XG. Hedyotis diffusa Willd extract inhibits the growth of human glioblastoma cells by inducing mitochondrial apoptosis via AKT/ERK pathways. JOURNAL OF ETHNOPHARMACOLOGY 2014; 158 Pt A:404-411. [PMID: 25456437 DOI: 10.1016/j.jep.2014.10.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 10/07/2014] [Accepted: 10/13/2014] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hedyotis diffusa Willd (Rubiaceae) (HDW) has been widely applied for the treatment of tumors, inflammation and toxication in traditional Chinese medicine. The antitumor effect of HDW on glioblastoma has been rarely reported. We aim to evaluate the activity of this extract and explore the underlying mechanism in U87 human glioblastoma cell line. MATERIALS AND METHODS Cytotoxicity of HDW extract on U87 cells was measured by MTT assay. Apoptosis, cell cycle arrest and mitochondrial membrane potential (MMP) collapse induced by HDW extract were determined by flow cytometry. Caspase activity was analyzed based on colorimetric assay with a microplate spectrophotometer. Protein expression was examined by Western blot. RESULTS HDW extract suppressed U87 cells growth in a dose- and time-dependent manner. Flow cytometry showed that HDW extract induced significant apoptosis, S/G2-M phase arrest and MMP collapse in U87 cells. Furthermore, dose-dependent activation of caspase-3, Bcl-2, Bax and ERK was observed with HDW extract treatment. Decreased Bcl-2/Bax ratio and Akt suppression were readily found as well. CONCLUSIONS Induction of mitochondria-mediated apoptosis played an essential role in antitumor activity of HDW extract in U87 cells, in which ERKs and Akt signaling proteins were also involved. These findings contributed to the feasibility of using HDW extract in glioblastoma treatment and the understanding of the molecular mechanism.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Neurosurgery, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Rui-Fan Xie
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qun-Gen Xiao
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ran Li
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao-Li Shen
- Department of Neurosurgery, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xin-Gen Zhu
- Department of Neurosurgery, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China.
| |
Collapse
|
37
|
Li G, Xie N, Yao Y, Zhang Y, Guo J, Feng Y, Lv F, Xiao RP, Cao CM. Identification of PI3K regulatory subunit p55γ as a novel inhibitor of vascular smooth muscle cell proliferation and neointimal formation. Cardiovasc Res 2014; 105:75-85. [PMID: 25388664 DOI: 10.1093/cvr/cvu235] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
AIMS Phosphatidylinositol 3 kinases (PI3Ks) play a pivotal role in vascular physiology and pathophysiology. We aimed to investigate the role of p55γ, a regulatory subunit of PI3Ks, in vascular smooth muscle cell (VSMC) proliferation and neointimal formation. METHODS AND RESULTS We identified p55γ as an important factor that suppresses VSMC proliferation and injury-evoked neointimal formation. Western blot and mRNA analyses showed that p55γ expression declined in balloon-injured rat carotid arteries and in response to PDGF-BB and serum treatment in cultured VSMCs. Overexpression of p55γ inhibited, whereas short hairpin RNA knockdown of p55γ promoted PDGF-BB- and serum-induced VSMC proliferation. Importantly, in vivo adenoviral gene transfer of p55γ into carotid arteries attenuated, while knockdown of p55γ enhanced balloon injury-induced neointimal formation. Furthermore, p55γ sequentially up-regulated p53 and p21, resulting in cell-cycle arrest in S phase; small-interfering RNA knockdown of either p53 or p21 blocked p55γ-induced VSMC growth arrest. Mechanistically, p55γ interacted with and stabilized p53 protein by blocking mouse double minute 2 homologue-mediated p53 ubiquitination and degradation, subsequently activating its target gene p21. Concurrently, p55γ up-regulated Bcl-xl expression, resulting in non-apoptotic growth arrest effect. CONCLUSION These findings mark p55γ as a novel upstream regulator of the p53-p21 signalling pathway that negatively regulates VSMC proliferation, suggesting that malfunction of p55γ may trigger vascular proliferative disorders.
Collapse
Affiliation(s)
- Geng Li
- Institute of Molecular Medicine, Peking University, Yiheyuan Road 5, Haidian District, Beijing 100871, China
| | - Ning Xie
- Institute of Molecular Medicine, Peking University, Yiheyuan Road 5, Haidian District, Beijing 100871, China
| | - Yuan Yao
- Institute of Molecular Medicine, Peking University, Yiheyuan Road 5, Haidian District, Beijing 100871, China
| | - Yan Zhang
- Institute of Molecular Medicine, Peking University, Yiheyuan Road 5, Haidian District, Beijing 100871, China
| | - Jiaojiao Guo
- Institute of Molecular Medicine, Peking University, Yiheyuan Road 5, Haidian District, Beijing 100871, China
| | - Yuanqing Feng
- Institute of Molecular Medicine, Peking University, Yiheyuan Road 5, Haidian District, Beijing 100871, China
| | - Fengxiang Lv
- Institute of Molecular Medicine, Peking University, Yiheyuan Road 5, Haidian District, Beijing 100871, China
| | - Rui-Ping Xiao
- Institute of Molecular Medicine, Peking University, Yiheyuan Road 5, Haidian District, Beijing 100871, China State Key Laboratory of Biomembrane and Membrane Biotechnology, Peking University, Beijing 100871, China Center for Life Sciences, Peking University, Yiheyuan Road 5, Haidian District, Beijing 100871, China
| | - Chun-Mei Cao
- Institute of Molecular Medicine, Peking University, Yiheyuan Road 5, Haidian District, Beijing 100871, China
| |
Collapse
|
38
|
Lin KW, Huang AM, Lin CC, Chang CC, Hsu WC, Hour TC, Pu YS, Lin CN. Anti-cancer effects of ursane triterpenoid as a single agent and in combination with cisplatin in bladder cancer. Eur J Pharmacol 2014; 740:742-51. [PMID: 24933647 DOI: 10.1016/j.ejphar.2014.05.051] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Revised: 05/01/2014] [Accepted: 05/07/2014] [Indexed: 01/06/2023]
Abstract
Ursolic acid and most of its derivatives are cytotoxic to bladder cancer cells. An ursolic acid derivative, isopropyl 3β-hydroxyurs-12-en-28-oat (UA17), previously reported that it exhibited potent cytotoxicity against bladder cancer cells, NTUB1 cells. In this study, we further investigated the underlying mechanism of UA17 and evaluated its potential clinical use. UA17 may exert the onset of a p53-mediated p38 MAPK activation to up-regulate GADD153. GADD153, in turn, down-regulated Bcl-2 protein to cause mitochondrial membrane potential loss and apoptosis through intracellular ROS generation. In addition, UA17 markedly decreased the levels of cyclins (D1 and E), cyclin-dependent kinases (CDK2 and CDK4), and caused increase of p21 and p27 levels. To assess the suitability of UA17 as a chemotherapeutic agent against NTUB1 cells, its cytotoxic effects have been further evaluated in the combination with cisplatin. The addition of UA17 to cisplatin induces possibly additive cell growth inhibition which correlated to the accumulation of S phase cells and a corresponding decrease in accumulation of G1 phase cells, accompanied an increased accumulation of sub-G1 phase cells. Furthermore, UA17/cisplatin combination exhibited increase of p21, cyclin E, and p-p53 level, and decrease of p27 and cyclin D1 proteins, and slightly diminishing the level of CDK2. P-p38 up-regulation induced by UA17/cisplatin combination through generation of ROS and Bcl-2 down-regulation induced by UA17/cisplatin combination increased cell death. Finally, the antitumorigenic effects of UA17 or UA17/cisplatin combination were further supported by their inhibition on growth of bladder tumor cells in a therapeutic murine MBT-2 bladder tumor model.
Collapse
Affiliation(s)
- Kai-Wei Lin
- Faculty of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - A-Mei Huang
- Department of Medicine, Graduate Institute of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Chi-Chen Lin
- Department of Medical Education and Research, Taichung Veterans General Hospital, Taichung 40705, Taiwan
| | - Chia-Che Chang
- Institute of Biomedical Sciences, National Chung Hsiung University, Taichung 40227, Taiwan
| | - Wei-Chi Hsu
- Department of Medicine, Graduate Institute of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Tzyh-Chyuan Hour
- Department of Medicine, Graduate Institute of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yeong-Shiau Pu
- Department of Urology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Chun-Nan Lin
- Faculty of Fragrance and Cosmetics, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Biological Science and Technology, School of Medicine, China Medical University, Taichung 40402, Taiwan.
| |
Collapse
|
39
|
Zhang K, Zhao X, Liu J, Fang X, Wang X, Wang X, Li R. β-diketone-cobalt complexes inhibit DNA synthesis and induce S-phase arrest in rat C6 glioma cells. Oncol Lett 2013; 7:881-885. [PMID: 24520304 PMCID: PMC3919926 DOI: 10.3892/ol.2013.1772] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 12/04/2013] [Indexed: 11/13/2022] Open
Abstract
β-diketone-cobalt complexes, a family of newly synthesized non-platinum metal compounds, exhibit potential antitumor activity; however, the antitumor mechanism is unclear. The current study investigated the mechanism by which β-diketone-cobalt complexes inhibit rat C6 glioma cell proliferation. It was found that β-diketone-cobalt complexes suppress rat C6 glioma cell viability in a dose-dependent manner (3.125–100 μg/ml). In rat C6 glioma cells, the IC50 value of β-diketone-cobalt complexes was 24.7±3.395 μg/ml and the IC10 value was 4.37±1.53 μg/ml, indicating a strong inhibitory effect. Further investigation suggested that β-diketone-cobalt complexes inhibit rat C6 glioma cell proliferation, which is associated with S-phase arrest and DNA synthesis inhibition. During this process, β-diketone-cobalt complexes decreased cyclin A expression and increased cyclin E and p21 expression. In addition, β-diketone-cobalt complexes exhibit a stronger antitumor capability than the antineoplastic agent, 5-fluorouracil.
Collapse
Affiliation(s)
- Kaizhi Zhang
- China-Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Xingli Zhao
- China-Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Junzhi Liu
- China-Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Xiangyang Fang
- China-Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Xuepeng Wang
- China-Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Xiaohong Wang
- Key Laboratory of Polyoxometalate Science of Ministry of Education, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Rui Li
- China-Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
40
|
De Maria S, Scognamiglio I, Lombardi A, Amodio N, Caraglia M, Cartenì M, Ravagnan G, Stiuso P. Polydatin, a natural precursor of resveratrol, induces cell cycle arrest and differentiation of human colorectal Caco-2 cell. J Transl Med 2013; 11:264. [PMID: 24138806 PMCID: PMC3854516 DOI: 10.1186/1479-5876-11-264] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 10/08/2013] [Indexed: 02/08/2023] Open
Abstract
Background Human colon adenocarcinoma cells are resistant to chemotherapeutic agents, such as anthracyclines, that induce death by increasing the reactive oxygen species. A number of studies have been focused on chemo-preventive use of resveratrol as antioxidant against cardiovascular diseases, aging and cancer. While resveratrol cytotoxic action was due to its pro-oxidant properties. In this study, we investigate whether the Resveratrol (trans-3,5,49-trihydroxystilbene) and its natural precursor Polydatin (resveratrol-3-O-b-mono- D-glucoside, the glycoside form of resveratrol) combination, might have a cooperative antitumor effect on either growing or differentiated human adenocarcinoma colon cancer cells. Methods The polydatin and resveratrol pharmacological interaction was evaluated in vitro on growing and differentiated Caco-2 cell lines by median drug effect analysis calculating a combination index with CalcuSyn software. We have selected a synergistic combination and we have evaluated its effect on the biological and molecular mechanisms of cell death. Results Simultaneous exposure to polydatin and resveratrol produced synergistic antiproliferative effects compared with single compound treatment. We demonstrated that polydatin alone or in combination with resveratrol at 3:1 molar ratio synergistically modulated oxidative stress, cell cycle, differentiation and apoptosis. Worthy of note treatment with polydatin induced a nuclear localization and decreased expression of heat shock protein 27, and vimentin redistributed within the cell. Conclusions From morphological, and biochemical outcome we obtained evidences that polydatin induced a transition from a proliferative morphology to cell-specific differentiated structures and caused human CaCo-2 cell death by induction of apoptosis. Our data suggest the potential use of polydatin in combination chemotherapy for human colon cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Paola Stiuso
- Department of Biochemistry, Biophisics and General Pathology, Second University of Naples, Naples, Italy.
| |
Collapse
|
41
|
Qiao S, Tao S, Rojo de la Vega M, Park SL, Vonderfecht AA, Jacobs SL, Zhang DD, Wondrak GT. The antimalarial amodiaquine causes autophagic-lysosomal and proliferative blockade sensitizing human melanoma cells to starvation- and chemotherapy-induced cell death. Autophagy 2013; 9:2087-102. [PMID: 24113242 DOI: 10.4161/auto.26506] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Pharmacological inhibition of autophagic-lysosomal function has recently emerged as a promising strategy for chemotherapeutic intervention targeting cancer cells. Repurposing approved and abandoned non-oncological drugs is an alternative approach to the identification and development of anticancer therapeutics, and antimalarials that target autophagic-lysosomal functions have recently attracted considerable attention as candidates for oncological repurposing. Since cumulative research suggests that dependence on autophagy represents a specific vulnerability of malignant melanoma cells, we screened a focused compound library of antimalarials for antimelanoma activity. Here we report for the first time that amodiaquine (AQ), a clinical 4-aminoquinoline antimalarial with unexplored cancer-directed chemotherapeutic potential, causes autophagic-lysosomal and proliferative blockade in melanoma cells that surpasses that of its parent compound chloroquine. Monitoring an established set of protein markers (LAMP1, LC3-II, SQSTM1) and cell ultrastructural changes detected by electron microscopy, we observed that AQ treatment caused autophagic-lysosomal blockade in malignant A375 melanoma cells, a finding substantiated by detection of rapid inactivation of lysosomal cathepsins (CTSB, CTSL, CTSD). AQ-treatment was associated with early induction of energy crisis (ATP depletion) and sensitized melanoma cells to either starvation- or chemotherapeutic agent-induced cell death. AQ displayed potent antiproliferative effects, and gene expression array analysis revealed changes at the mRNA (CDKN1A, E2F1) and protein level (TP53, CDKN1A, CCND1, phospho-RB1 [Ser 780]/[Ser 807/811], E2F1) consistent with the observed proliferative blockade in S-phase. Taken together, our data suggest that the clinical antimalarial AQ is a promising candidate for repurposing efforts that aim at targeting autophagic-lysosomal function and proliferative control in malignant melanoma cells.
Collapse
Affiliation(s)
- Shuxi Qiao
- Department of Pharmacology and Toxicology; College of Pharmacy and Arizona Cancer Center; University of Arizona; Tucson, AZ USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Wu M, Zhang H, Hu J, Weng Z, Li C, Li H, Zhao Y, Mei X, Ren F, Li L. Isoalantolactone inhibits UM-SCC-10A cell growth via cell cycle arrest and apoptosis induction. PLoS One 2013; 8:e76000. [PMID: 24098753 PMCID: PMC3786891 DOI: 10.1371/journal.pone.0076000] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 08/23/2013] [Indexed: 12/03/2022] Open
Abstract
Isoalantolactone is a sesquiterpene lactone compound isolated from the roots of Inula helenium L. Previous studies have demonstrated that isoalantolactone possesses antifungal, anti-bacterial, anti-helminthic and anti-proliferative properties in a variety of cells, but there are no studies concerning its effects on head and neck squamous cell carcinoma (HNSCC). In the present study, an MTT assay demonstrated that isoalantolactone has anti-proliferative activity against the HNSCC cell line (UM-SCC-10A). Immunostaining identified that this compound induced UM-SCC-10A cell apoptosis but not necrosis. To explain the molecular mechanisms underlying its effects, flow cytometry and western blot analysis showed that the apoptosis was associated with cell cycle arrest during the G1 phase, up-regulation of p53 and p21, and down-regulation of cyclin D. Furthermore, our results revealed that induction of apoptosis through a mitochondrial pathway led to up-regulation of pro-apoptotic protein expression (Bax), down-regulation of anti-apoptotic protein expression (Bcl-2), mitochondrial release of cytochrome c (Cyto c), reduction of mitochondrial membrane potential (MMP) and activation of caspase-3 (Casp-3). Involvement of the caspase apoptosis pathway was confirmed using caspase inhibitor Z-VAD-FMK pretreatment. Together, our findings suggest that isoalantolactone induced caspase-dependent apoptosis via a mitochondrial pathway and was associated with cell cycle arrest in the G1 phase in UM-SCC-10A cells. Therefore, isoalantolactone may become a potential drug for treating HNSCC.
Collapse
Affiliation(s)
- Minjun Wu
- Department of Cell Biology and Anatomy, Liaoning Medical University, Jinzhou, China
| | - Hua Zhang
- Department of Cell Biology and Anatomy, Liaoning Medical University, Jinzhou, China
| | - Jiehua Hu
- Educational Technologies and Simulation Training Centre, Naval University of Engineering Tianjin Campus, Tianjin, China
| | - Zhiyong Weng
- Department of Cell Biology and Anatomy, Liaoning Medical University, Jinzhou, China
| | - Chenyuan Li
- Department of Cell Biology and Anatomy, Liaoning Medical University, Jinzhou, China
| | - Hong Li
- Department of Cell Biology and Anatomy, Liaoning Medical University, Jinzhou, China
| | - Yan Zhao
- Department of Cell Biology and Anatomy, Liaoning Medical University, Jinzhou, China
| | - Xifan Mei
- Department of Cell Biology and Anatomy, Liaoning Medical University, Jinzhou, China
| | - Fu Ren
- Department of Cell Biology and Anatomy, Liaoning Medical University, Jinzhou, China
- * E-mail: (FR); (LL)
| | - Lihua Li
- Department of Cell Biology and Anatomy, Liaoning Medical University, Jinzhou, China
- * E-mail: (FR); (LL)
| |
Collapse
|
43
|
Simvastatin induces caspase-dependent apoptosis and activates P53 in OCM-1 cells. Exp Eye Res 2013; 113:128-34. [PMID: 23727160 DOI: 10.1016/j.exer.2013.05.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2013] [Revised: 05/05/2013] [Accepted: 05/17/2013] [Indexed: 11/23/2022]
Abstract
Simvastatin is a cholesterol-lowering drug which exhibits numerous pleiotropic effects including anti-cancer activity. Yet, the anti-cancer effects in choroidal melanoma remain poorly characterized. Therefore, in this study, we investigated the effects of simvastatin on OCM-1 cells growth, apoptosis and cycle. Simvastatin showed an inhibitory effects on OCM-1 cells viability in dose-dependent (2-10 μM) and time-dependent (24-72 h) manner. Further study suggested that simvastatin-induced inhibition OCM-1 cells proliferation was associated with G1 phase arrest, decreased protein and mRNA expression of proliferation marker cyclin D1, cyclin E, cyclin dependent kinase (CDK)2 and increased expression of CDK inhibitory protein P21. In addition, simvastatin resulted in an increase in levels of reactive oxygen species (ROS) in OCM-1 cells and simvastatin significantly triggered apoptosis in OCM-1 cells, which was characterized by increased chromatin condensation, activation of caspase-9 and cleaved-caspase-3, increased expression mitochondrion-related apoptosis protein of P53, Bax and decreased expression of Bcl2 and iASPP. Collectively, our study demonstrated that simvastatin can efficiently inhibit proliferation and induce apoptosis in OCM-1 cells.
Collapse
|
44
|
Okochi-Takada E, Hattori N, Tsukamoto T, Miyamoto K, Ando T, Ito S, Yamamura Y, Wakabayashi M, Nobeyama Y, Ushijima T. ANGPTL4 is a secreted tumor suppressor that inhibits angiogenesis. Oncogene 2013; 33:2273-8. [PMID: 23686315 DOI: 10.1038/onc.2013.174] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 02/14/2013] [Accepted: 03/28/2013] [Indexed: 12/12/2022]
Abstract
Tumor suppressors with extracellular function are likely to have advantages as targets for cancer therapy, but few are known. Here, we focused on angiopoietin-like 4 (ANGPTL4), which is a secreted glycoprotein involved in lipoprotein metabolism and angiogenesis, is methylation-silenced in human cancers, but has unclear roles in cancer development and progression. We found a deletion mutation in its coiled-coil domain at its N-terminal in human gastric cancers, in addition to hypermethylation of the ANGPTL4 promoter CpG islands. Forced expression of wild-type ANGPTL4, but not ANGPTL4 with the deletion, at physiological levels markedly suppressed in vivo tumorigenicity and tumor angiogenesis, indicating that the latter caused the former. Tumor-derived ANGPTL4 suppressed in vitro vascular tube formation and proliferation of human umbilical vascular endothelial cells, partly due to suppression of ERK signaling. These showed that ANGPTL4 is a genetically and epigenetically inactivated secreted tumor suppressor that inhibits tumor angiogenesis.
Collapse
Affiliation(s)
- E Okochi-Takada
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - N Hattori
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - T Tsukamoto
- Oncological Pathology Division, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - K Miyamoto
- Division of Molecular Oncology, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Kureshi, Japan
| | - T Ando
- 1] Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan [2] Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - S Ito
- Department of Gastroenterological Surgery, Aichi Cancer Center Central Hospital, Nagoya, Japan
| | - Y Yamamura
- Department of Gastroenterological Surgery, Aichi Cancer Center Central Hospital, Nagoya, Japan
| | - M Wakabayashi
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Y Nobeyama
- 1] Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan [2] Department of Dermatology, The Jikei University School of Medicine, Tokyo, Japan
| | - T Ushijima
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
45
|
O'Neal MA, Posner BA, Coates CJ, Abrams JM. A cell-based screening platform identifies novel mosquitocidal toxins. ACTA ACUST UNITED AC 2013; 18:688-94. [PMID: 23407365 DOI: 10.1177/1087057113476952] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Pesticides currently in widespread use often lack species specificity and also become less effective as resistance emerges. Consequently, there is a pressing need to develop novel agents that are narrowly targeted and safe to humans. A cell-based screening platform was designed to discover compounds that are lethal to mosquito (Anopheles and Aedes) cells but show little or no activity against other insect (Drosophila) or human cell lines. Mosquito-specific, aqueous-stable cytotoxins were recovered at rare frequencies. Three of these were profiled for structure-activity relationships and also assessed in whole-animal toxicity assays. In at least one test case, species-specific cytotoxicity seen in culture effectively translated to the whole-animal level, with potent toxicity against Anopheles yet none against Drosophila. Therefore, this initiative has the potential to advance novel mosquitocidal agents and, in a broader sense, could establish a versatile platform for developing customized pesticides that selectively target other disease vectors as well.
Collapse
Affiliation(s)
- Melissa A O'Neal
- University of Texas Southwestern Medical Center, Department of Cell Biology, Dallas, TX, USA
| | | | | | | |
Collapse
|
46
|
Yao Y, Zhang YW, Sun LG, Liu B, Bao YL, Lin H, Zhang Y, Zheng LH, Sun Y, Yu CL, Wu Y, Wang GN, Li YX. Juglanthraquinone C, a novel natural compound derived from Juglans mandshurica Maxim, induces S phase arrest and apoptosis in HepG2 cells. Apoptosis 2012; 17:832-41. [PMID: 22484481 DOI: 10.1007/s10495-012-0722-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Juglanthraquinone C (1,5-dihydroxy-9,10-anthraquinone-3-carboxylic acid, JC), a naturally occurring anthraquinone isolated from the stem bark of Juglans mandshurica, shows strong cytotoxicity in various human cancer cells in vitro. Here, we first performed a structure-activity relationship study of six anthraquinone compounds (JC, rhein, emodin, aloe-emodin, physcion and chrysophanol) to exploit the relationship between their structural features and activity. The results showed that JC exhibited the strongest cytotoxicity of all compounds evaluated. Next, we used JC to treat several human cancer cell lines and found that JC showed an inhibitory effect on cell viability in dose-dependent (2.5-10 μg/ml JC) and time-dependent (24-48 h) manners. Importantly, the inhibitory effect of JC on HepG2 (human hepatocellular carcinoma) cells was more significant as shown by an IC(50) value of 9 ± 1.4 μg/ml, and 36 ± 1.2 μg/ml in L02 (human normal liver) cells. Further study suggested that JC-induced inhibition HepG2 cell proliferation was associated with S phase arrest, decreased protein expression of proliferation marker Ki67, cyclin A and cyclin-dependent kinase (CDK) 2, and increased expression of cyclin E and CDK inhibitory protein Cip1/p21. In addition, JC significantly triggered apoptosis in HepG2 cells, which was characterized by increased chromatin condensation and DNA fragmentation, activation of caspase-9 and -3, and induction of a higher Bax/Bcl2 ratio. Collectively, our study demonstrated that JC can efficiently inhibit proliferation and induce apoptosis in HepG2 cells.
Collapse
Affiliation(s)
- Yao Yao
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lin YJ, Zhen YZ, Zhao YF, Wei J, Hu G. Rhein Lysinate Induced S-Phase Arrest and Increased the Anti-Tumor Activity of 5-FU in HeLa Cells. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2012; 39:817-25. [PMID: 21721159 DOI: 10.1142/s0192415x11009226] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Rhein lysinate (RHL), easily dissolved in water, is one of the anthraquinones, and has been shown to have anti-tumor activity in different human cancer cell lines. In the present study, we observed that RHL could cause vacuolar degeneration in HeLa cells, which was not observed in human umbilical vein endothelial cells (HUVECs) and other cell lines (SKOV-3 and SK-BR-3). Therefore, the purpose of this study was to investigate the anti-tumor effect of rhein lysinate on human cervix cancer HeLa cells. The results indicated that RHL could induce HeLa cell S-phase arrest and RHL (higher than 80 μM) also induced HeLa cell G2/M-phase arrest in a dose-dependent manner. Compared to the HeLa cells, RHL induced HUVECs G1-phase arrest at all dose levels tested in a dose-dependent manner. Treatment with RHL led to a significant S or G2/M-phase arrest through promoting the expression of p53 and p21 and the phosphorylation of p53. Moreover, 80 μM RHL could increase 5-FU anti-tumor activity. In conclusion, RHL could be a novel chemotherapeutic drug candidate for the treatment of human cervix cancer in the future.
Collapse
Affiliation(s)
- Ya-Jun Lin
- Beijing Hospital and Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, P. R. China
| | - Yong-Zhan Zhen
- Basic Medical College, Hebei United University, Tangshan, Hebei, P. R. China
| | - Yu-Fang Zhao
- Basic Medical College, Hebei United University, Tangshan, Hebei, P. R. China
| | - Jie Wei
- Beijing Hospital and Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, P. R. China
| | - Gang Hu
- Beijing Hospital and Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, P. R. China
| |
Collapse
|
48
|
Komarasamy TV, Sekaran SD. The anti-proliferative effects of a palm oil-derived product and its mode of actions in human malignant melanoma MeWo cells. J Oleo Sci 2012; 61:227-39. [PMID: 22450124 DOI: 10.5650/jos.61.227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Melanoma incidence and mortality have risen dramatically in recent years. No effective treatment for metastatic melanoma exists; hence currently, an intense effort for new drug evaluation is being carried out. In this study, we investigated the effects of a palm oil-derived nanopolymer called Bio-12 against human malignant melanoma. The nanopolymers of Bio-12 are lipid esters derived from a range of fatty acids of palm oil. Our study aims to identify the anti-proliferative properties of Bio-12 against human malignant melanoma cell line (MeWo) and to elucidate the mode of actions whereby Bio-12 brings about cell death. Bio-12 significantly inhibited the growth of MeWo cells in a concentration- and time- dependent manner with a median inhibitory concentration (IC₅₀) value of 1/25 dilution after 72 h but was ineffective on human normal skin fibroblasts (CCD-1059sk). We further investigated the mode of actions of Bio-12 on MeWo cells. Cell cycle flow cytometry demonstrated that MeWo cells treated with increasing concentrations of Bio-12 resulted in S-phase arrest, accompanied by the detection of sub-G1 content, indicative of apoptotic cell death. Induction of apoptosis was further confirmed via caspase (substrate) cleavage assay which showed induction of early apoptosis in MeWo cells. In addition, DNA strand breaks which are terminal event in apoptosis were evident through increase of TUNEL positive cells and formation of a characteristic DNA ladder on agarose gel electrophoresis. Moreover, treatment of MeWo cells with Bio-12 induced significant increase in lactate dehydrogenase (LDH) activity. These results show that Bio-12 possesses the ability to suppress proliferation of human malignant melanoma MeWo cells and this suppression is at least partly attributed to the initiation of the S-phase arrest, apoptosis and necrosis, suggesting that it is indeed worth for further investigations.
Collapse
Affiliation(s)
- Thamil Vaani Komarasamy
- Department of Medical Microbiology, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
| | | |
Collapse
|
49
|
An alternative Kaposi's sarcoma-associated herpesvirus replication program triggered by host cell apoptosis. J Virol 2012; 86:4404-19. [PMID: 22345480 DOI: 10.1128/jvi.06617-11] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is linked to several neoplastic diseases: Kaposi's sarcoma, primary effusion lymphoma (PEL), and multicentric Castleman's disease (MCD). KSHV replicates actively, via a controlled gene expression program, but can also remain latent. It had been thought that the transition from latent to lytic replication was controlled exclusively by the replication and transcription activator protein RTA (open reading frame 50 [ORF50] gene product). A dominant-negative (DN) ORF50 mutant, ORF50ΔSTAD, blocks gene expression and replication. We produced a PEL cell line derivative containing both latent KSHV genomes and an inducible ORF50ΔSTAD. We unexpectedly found that induction of apoptosis triggered high-level viral replication, even when DN ORF50ΔSTAD was present, suggesting that apoptosis triggers KSHV replication through a distinct RTA-independent pathway. We verified that apoptosis triggers KSHV replication independent of RTA using ORF50 small interfering RNA (siRNA) and also showed that caspase activity is required to trigger KSHV replication. We showed that when apoptosis triggers KSHV replication, the kinetics of late gene expression is accelerated by 12 to 24 h and that virus produced following apoptosis has reduced infectivity. KSHV therefore appears to replicate via two distinct pathways, a conventional pathway requiring RTA, with slower replication kinetics, producing virus with higher infectivity, and an alternative apoptosis-triggered pathway that does not require RTA, has faster replication kinetics, and produces virus with lower infectivity. The existence of a distinct apoptosis-triggered, accelerated replication pathway may have evolutionary advantages for the virus and clinical significance for the treatment of KSHV-associated neoplasms. It also provides further evidence that KSHV can sense and react to its environment.
Collapse
|
50
|
Liu W, Chen Y, Lu G, Sun L, Si J. Down-regulation of HSP70 sensitizes gastric epithelial cells to apoptosis and growth retardation triggered by H. pylori. BMC Gastroenterol 2011; 11:146. [PMID: 22208848 PMCID: PMC3264526 DOI: 10.1186/1471-230x-11-146] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Accepted: 12/30/2011] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND H. pylori infection significantly attenuated the expression of HSP70 in gastric mucosal cells. However, the role of HSP70 cancellation in H. pylori-associated cell damages is largely unclear. METHODS Small interfering RNA (siRNA) was used to down-regulate HSP70 in gastric epithelial cell lines AGS. The transfected cells were then incubated with H. pylori and the functions of HSP70 suppression were observed by viability assay, cell cycle analyses and TUNEL assay. HSP70 target apoptotic proteins were further identified by Western blot. RESULTS The inhibition of HSP70 has further increased the effect of growth arrest and apoptosis activation triggered by H. pylori in gastric epithelial cells. The anti-proliferation function of HSP70 depletion was at least by up-regulating p21 and cell cycle modulation with S-phase accumulation. An increase of apoptosis-inducing factor (AIF) and cytosolic cytochrome C contributes to the activation of apoptosis following down-regulation of intracellular HSP70. Extracellular HSP70 increased cellular resistance to apoptosis by suppression the release of AIF and cytochrome c from mitochondria, as well as inhibition of p21 expression. CONCLUSIONS The inhibition of HSP70 aggravated gastric cellular damages induced by H. pylori. Induction of HSP70 could be a potential therapeutic target for protection gastric mucosa from H. pylori-associated injury.
Collapse
Affiliation(s)
- Weili Liu
- Gastroenterology laboratory, Clinical Research Institute, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 310016 Hangzhou, People's Republic of China
| | | | | | | | | |
Collapse
|