1
|
Yaghmur A, Østergaard J, Mu H. Lipid nanoparticles for targeted delivery of anticancer therapeutics: Recent advances in development of siRNA and lipoprotein-mimicking nanocarriers. Adv Drug Deliv Rev 2023; 203:115136. [PMID: 37944644 DOI: 10.1016/j.addr.2023.115136] [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] [Received: 08/31/2023] [Revised: 10/19/2023] [Accepted: 11/04/2023] [Indexed: 11/12/2023]
Abstract
The limitations inherent in conventional cancer treatment methods have stimulated recent efforts towards the design of safe nanomedicines with high efficacy for combating cancer through various promising approaches. A plethora of nanoparticles has been introduced in the development of cancer nanomedicines. Among them, different lipid nanoparticles are attractive for use due to numerous advantages and unique opportunities, including biocompatibility and targeted drug delivery. However, a comprehensive understanding of nano-bio interactions is imperative to facilitate the translation of recent advancements in the development of cancer nanomedicines into clinical practice. In this contribution, we focus on lipoprotein-mimicking nanoparticles, which possess unique features and compositions facilitating drug transport through receptor binding mechanisms. Additionally, we describe potential applications of siRNA lipid nanoparticles in the future design of anticancer nanomedicines. Thus, this review highlights recent progress, challenges, and opportunities of lipid-based lipoprotein-mimicking nanoparticles and siRNA nanocarriers designed for the targeted delivery of anticancer therapeutic agents.
Collapse
Affiliation(s)
- Anan Yaghmur
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Jesper Østergaard
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Huiling Mu
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| |
Collapse
|
2
|
Ebid H, Trombetta LD. Effects of glyphosate, mancozeb and their combinations on mouse neuroblastoma cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 104:104302. [PMID: 37871707 DOI: 10.1016/j.etap.2023.104302] [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: 08/01/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 10/25/2023]
Abstract
Pesticides-related toxicities have long been studied. Data regarding the effects of combined exposure to environmentally relevant pesticides however remain lacking. The herbicide glyphosate and the fungicide mancozeb are extensively used in agriculture. Residues of both compounds are frequently found in food and water and therefore, environmental exposure to both pesticides is a possibility. Neurotoxicity of glyphosate, mancozeb and their combinations were investigated using mouse neuroblastoma cells. Cytotoxicity observed with the glyphosate and mancozeb combinations was higher than that observed when glyphosate was tested alone. Combinations of glyphosate followed by mancozeb increased copper, manganese, and zinc levels. Mixture of mancozeb + glyphosate increased manganese and zinc levels. Combination of mancozeb followed by glyphosate increased copper and zinc levels. Glutathione ratio was decreased as a result of combinations of glyphosate and mancozeb. The decrease in glutathione ratio was greater in the combination groups than in glyphosate alone.
Collapse
Affiliation(s)
- Heidi Ebid
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Jamaica, NY 11439, USA
| | - Louis D Trombetta
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Jamaica, NY 11439, USA.
| |
Collapse
|
3
|
Kaur P, Sharma S, Goel A, Sharma P, Agnihotri N, Kaur R, Singh V. 4‐Hydroxy Enigmol, a 1‐Deoxyphytosphingolipid that Exhibit Good Activity against Prostate and Colon Cancer. ChemistrySelect 2023. [DOI: 10.1002/slct.202203861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Parleen Kaur
- Department of Applied Scienced Punjab Engineering College Deemed to be University) 160 012 Chandigarh India
| | - Sonia Sharma
- Department cum National Genomics studies and Research Panjab University 160 014 Chandigarh India
| | - Akshita Goel
- Department of Chemistry and Centre of Advanced Studies in Chemistry Panjab University 160 014 Chandigarh India
| | - Purshotam Sharma
- Department of Chemistry and Centre of Advanced Studies in Chemistry Panjab University 160 014 Chandigarh India
| | - Navneet Agnihotri
- Department of biochemistry Panjab University 160 025 Chandigarh India
| | - Ramandeep Kaur
- Department cum National Genomics studies and Research Panjab University 160 014 Chandigarh India
| | - Vasundhara Singh
- Department of Applied Scienced Punjab Engineering College Deemed to be University) 160 012 Chandigarh India
| |
Collapse
|
4
|
Chaudhry GES, Md Akim A, Sung YY, Sifzizul TMT. Cancer and apoptosis: The apoptotic activity of plant and marine natural products and their potential as targeted cancer therapeutics. Front Pharmacol 2022; 13:842376. [PMID: 36034846 PMCID: PMC9399632 DOI: 10.3389/fphar.2022.842376] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 07/13/2022] [Indexed: 11/24/2022] Open
Abstract
Cancer is a multifactorial, multi-stage disease, including complex cascades of signaling pathways—the cell growth governed by dysregulated and abrupt cell division. Due to the complexity and multi-regulatory cancer progression, cancer is still a challenging disease to treat and survive. The screening of extracts and fractions from plants and marine species might lead to the discovery of more effective compounds for cancer therapeutics. The isolated compounds and reformed analogs were known as future prospective contenders for anti-cancer chemotherapy. For example, Taxol, a potent mitotic inhibitor discovered from Taxus brevifolia, suppresses cell growth and arrest, induces apoptosis, and inhibits proliferation. Similarly, marine sponges show remarkable tumor chemo preventive and chemotherapeutic potential. However, there is limited research to date. Several plants and marine-derived anti-cancer compounds having the property to induce apoptosis have been approved for clinical trials. The anti-cancer activity kills the cell and slows the growth of cancer cells. Among cell death mechanisms, apoptosis induction is a more profound mechanism of cell death triggered by naturally isolated anti-cancer agents. Evading apoptosis is the major hurdle in killing cancer cells, a mechanism mainly regulated as intrinsic and extrinsic. However, it is possible to modify the apoptosis-resistant phenotype of the cell by altering many of these mechanisms. Various extracts and fractions successfully induce apoptosis, cell-cycle modulation, apoptosis, and anti-proliferative activity. Therefore, there is a pressing need to develop new anti-cancer drugs of natural origins to reduce the effects on normal cells. Here, we’ve emphasized the most critical elements: i) A better understanding of cancer progression and development and its origins, ii) Molecular strategies to inhibit the cell proliferation/Carcino-genesis, iii) Critical regulators of cancer cell proliferation and development, iv) Signaling Pathways in Apoptosis: Potential Targets for targeted therapeutics, v) Why Apoptosis induction is mandatory for effective chemotherapy, vi) Plants extracts/fractions as potential apoptotic inducers, vii) Marine extracts as Apoptotic inducers, viii) Marine isolated Targeted compounds as Apoptotic inducers (FDA Approved/treatment Phase). This study provides a potential therapeutic option for cancer, although more clinical studies are needed to verify its efficacy in cancer chemotherapy.
Collapse
Affiliation(s)
- Gul-e-Saba Chaudhry
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Terengganu, Malaysia
- *Correspondence: Gul-e-Saba Chaudhry, ,
| | - Abdah Md Akim
- Department of Biomedical Sciences, Faculty of Medicine and Health sciences, University of Putra Malaysia, Seri Kembangan, Malaysia
| | - Yeong Yik Sung
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Terengganu, Malaysia
| | | |
Collapse
|
5
|
Michala AS, Pritsa A. Quercetin: A Molecule of Great Biochemical and Clinical Value and Its Beneficial Effect on Diabetes and Cancer. Diseases 2022; 10:37. [PMID: 35892731 PMCID: PMC9326669 DOI: 10.3390/diseases10030037] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 02/01/2023] Open
Abstract
Quercetin belongs to the broader category of polyphenols. It is found, in particular, among the flavonols, and along with kaempferol, myricetin and isorhamnetin, it is recognized as a foreign substance after ingestion in contrast to vitamins. Quercetin occurs mainly linked to sugars with the most common compounds being quercetin-3-O-glucoside or as an aglycone, especially in the plant population. The aim of this review is to present a recent bibliography on the mechanisms of quercetin absorption and metabolism, bioavailability, and antioxidant and the clinical effects in diabetes and cancer. The literature reports a positive effect of quercetin on oxidative stress, cancer, and the regulation of blood sugar levels. Moreover, research-administered drug dosages of up to 2000 mg per day showed mild to no symptoms of overdose. It should be noted that quercetin is no longer considered a carcinogenic substance. The daily intake of quercetin in the diet ranges 10 mg-500 mg, depending on the type of products consumed. This review highlights that quercetin is a valuable dietary antioxidant, although a specific daily recommended intake for this substance has not yet been determined and further studies are required to decide a beneficial concentration threshold.
Collapse
Affiliation(s)
| | - Agathi Pritsa
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University (IHU), P.O. 141 Sindos, 57400 Thessaloniki, Greece;
| |
Collapse
|
6
|
Guevara L, Domínguez-Anaya MÁ, Ortigosa A, González-Gordo S, Díaz C, Vicente F, Corpas FJ, Pérez del Palacio J, Palma JM. Identification of Compounds with Potential Therapeutic Uses from Sweet Pepper ( Capsicum annuum L.) Fruits and Their Modulation by Nitric Oxide (NO). Int J Mol Sci 2021; 22:ijms22094476. [PMID: 33922964 PMCID: PMC8123290 DOI: 10.3390/ijms22094476] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/15/2021] [Accepted: 04/20/2021] [Indexed: 12/14/2022] Open
Abstract
Plant species are precursors of a wide variety of secondary metabolites that, besides being useful for themselves, can also be used by humans for their consumption and economic benefit. Pepper (Capsicum annuum L.) fruit is not only a common food and spice source, it also stands out for containing high amounts of antioxidants (such as vitamins C and A), polyphenols and capsaicinoids. Particular attention has been paid to capsaicin, whose anti-inflammatory, antiproliferative and analgesic activities have been reported in the literature. Due to the potential interest in pepper metabolites for human use, in this project, we carried out an investigation to identify new bioactive compounds of this crop. To achieve this, we applied a metabolomic approach, using an HPLC (high-performance liquid chromatography) separative technique coupled to metabolite identification by high resolution mass spectrometry (HRMS). After chromatographic analysis and data processing against metabolic databases, 12 differential bioactive compounds were identified in sweet pepper fruits, including quercetin and its derivatives, L-tryptophan, phytosphingosin, FAD, gingerglycolipid A, tetrahydropentoxylin, blumenol C glucoside, colnelenic acid and capsoside A. The abundance of these metabolites varied depending on the ripening stage of the fruits, either immature green or ripe red. We also studied the variation of these 12 metabolites upon treatment with exogenous nitric oxide (NO), a free radical gas involved in a good number of physiological processes in higher plants such as germination, growth, flowering, senescence, and fruit ripening, among others. Overall, it was found that the content of the analyzed metabolites depended on the ripening stage and on the presence of NO. The metabolic pattern followed by quercetin and its derivatives, as a consequence of the ripening stage and NO treatment, was also corroborated by transcriptomic analysis of genes involved in the synthesis of these compounds. This opens new research perspectives on the pepper fruit’s bioactive compounds with nutraceutical potentiality, where biotechnological strategies can be applied for optimizing the level of these beneficial compounds.
Collapse
Affiliation(s)
- Lucía Guevara
- Group of Antioxidant, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, 18008 Granada, Spain; (L.G.); (M.Á.D.-A.); (A.O.); (S.G.-G.); (F.J.C.)
| | - María Ángeles Domínguez-Anaya
- Group of Antioxidant, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, 18008 Granada, Spain; (L.G.); (M.Á.D.-A.); (A.O.); (S.G.-G.); (F.J.C.)
| | - Alba Ortigosa
- Group of Antioxidant, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, 18008 Granada, Spain; (L.G.); (M.Á.D.-A.); (A.O.); (S.G.-G.); (F.J.C.)
| | - Salvador González-Gordo
- Group of Antioxidant, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, 18008 Granada, Spain; (L.G.); (M.Á.D.-A.); (A.O.); (S.G.-G.); (F.J.C.)
| | - Caridad Díaz
- Department of Screening & Target Validation, Fundación MEDINA, 18016 Granada, Spain; (C.D.); (F.V.); (J.P.d.P.)
| | - Francisca Vicente
- Department of Screening & Target Validation, Fundación MEDINA, 18016 Granada, Spain; (C.D.); (F.V.); (J.P.d.P.)
| | - Francisco J. Corpas
- Group of Antioxidant, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, 18008 Granada, Spain; (L.G.); (M.Á.D.-A.); (A.O.); (S.G.-G.); (F.J.C.)
| | - José Pérez del Palacio
- Department of Screening & Target Validation, Fundación MEDINA, 18016 Granada, Spain; (C.D.); (F.V.); (J.P.d.P.)
| | - José M. Palma
- Group of Antioxidant, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, 18008 Granada, Spain; (L.G.); (M.Á.D.-A.); (A.O.); (S.G.-G.); (F.J.C.)
- Correspondence: ; Tel.: +34-958-181-1600; Fax: +34-958-181-609
| |
Collapse
|
7
|
The Most Competent Plant-Derived Natural Products for Targeting Apoptosis in Cancer Therapy. Biomolecules 2021; 11:biom11040534. [PMID: 33916780 PMCID: PMC8066452 DOI: 10.3390/biom11040534] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/17/2021] [Accepted: 03/31/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer is a challenging problem for the global health community, and its increasing burden necessitates seeking novel and alternative therapies. Most cancers share six basic characteristics known as "cancer hallmarks", including uncontrolled proliferation, refractoriness to proliferation blockers, escaping apoptosis, unlimited proliferation, enhanced angiogenesis, and metastatic spread. Apoptosis, as one of the best-known programmed cell death processes, is generally promoted through two signaling pathways, including the intrinsic and extrinsic cascades. These pathways comprise several components that their alterations can render an apoptosis-resistance phenotype to the cell. Therefore, targeting more than one molecule in apoptotic pathways can be a novel and efficient approach for both identifying new anticancer therapeutics and preventing resistance to therapy. The main purpose of this review is to summarize data showing that various plant extracts and plant-derived molecules can activate both intrinsic and extrinsic apoptosis pathways in human cancer cells, making them attractive candidates in cancer treatment.
Collapse
|
8
|
Kim TW, Hong DW, Park JW, Hong SH. CB11, a novel purine-based PPARɣ ligand, overcomes radio-resistance by regulating ATM signalling and EMT in human non-small-cell lung cancer cells. Br J Cancer 2020; 123:1737-1748. [PMID: 32958825 PMCID: PMC7723055 DOI: 10.1038/s41416-020-01088-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 07/27/2020] [Accepted: 09/02/2020] [Indexed: 01/03/2023] Open
Abstract
Background Peroxisome proliferator-activated receptor γ (PPARγ) agonists frequently induce cell death in human non-small-cell lung cancer (NSCLC) cells. However, majority of NSCLC patients acquire resistance after cancer therapy, and it is still unclear. Methods In this study we investigated the apoptotic mechanism and the anti-cancer effects of a novel purine-based PPARγ agonist, CB11 (8-(2-aminophenyl)-3-butyl-1,6,7-trimethyl-1H-imidazo[2,1-f]purine-2,4(3H,8H)-dione), on human NSCLC cells. CB11 mediates PPARγ-dependent cell death, reactive oxygen species (ROS) production, mitochondrial membrane potential (MMP) collapse, cell cycle arrest, lactate dehydrogenase (LDH) cytotoxicity, and caspase-3 activity in human NSCLC cells. Results CB11 causes cell death via ROS-mediated ATM-p53-GADD45α signalling in human NSCLC cells, and diphenyleneiodonium (DPI), an NADPH oxidase inhibitor, decreases cell death by inhibiting CB11-mediated ATM signalling. In a xenograft experiment, CB11 dramatically reduced tumour volume when compared to a control group. Furthermore, CB11 induced cell death by inhibiting epithelial-to-mesenchymal transition (EMT) under radiation exposure in radiation-resistant human NSCLC cells. However, PPARγ deficiency inhibited cell death by blocking the ATM-p53 axis in radiation/CB11-induced radiation-resistant human NSCLC cells. Conclusions Taken together, our results suggest that CB11, a novel PPARγ agonist, may be a novel anti-cancer agent, and it could be useful in a therapeutic strategy to overcome radio-resistance in radiation-exposed NSCLC.
Collapse
Affiliation(s)
- Tae Woo Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, 139-706, Republic of Korea
| | - Da-Won Hong
- Laboratory of RNA Cell Biology, Graduate Department of Bioconvergence Science and Technology, Dankook University, Jukjeon-ro 152, Suji-gu, Yongin-si, Gyeonggi-do, 16892, Republic of Korea
| | - Joung Whan Park
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, 139-706, Republic of Korea
| | - Sung Hee Hong
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, 139-706, Republic of Korea.
| |
Collapse
|
9
|
Erekat NS. Cerebellar Upregulation of Cell Surface Death Receptor-Mediated Apoptotic Factors in Harmaline-Induced Tremor: An Immunohistochemistry Study. J Cell Death 2018; 11:1179066018809091. [PMID: 30450003 PMCID: PMC6236486 DOI: 10.1177/1179066018809091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 09/30/2018] [Indexed: 11/24/2022] Open
Abstract
Active caspase-3-mediated apoptosis has been implicated in the pathogenesis of
harmaline-induced tremor. The aim of this study is to illustrate the impact of
tremor induction on the expression of factors mediating the cell surface death
receptor–dependent apoptosis. A total of 20 normal Wistar rats were randomly
selected and equally divided into control and experimental groups. Tremor was
induced in the experimental group by injecting the rats with a single dose of
harmaline (50 mg/kg). After that, cerebellar tissues were evaluated by
immunohistochemistry to examine the expression of tumor necrosis factor α
(TNF-α) and active caspase-8 in the 2 groups of animals. TNF-α and active
caspase-8 expression was significantly higher in cerebella from experimental
rats compared with that in those from the control rats (P
value < .01). Thus, our present data suggest the association of tremor
induction with the cerebellar overexpression of TNF-α and active caspase-8,
correlative with Purkinje cell (PC) loss indicated by loss of calbindin
immunoreactivity, indicating the induction of the cell surface death
receptor–mediated apoptosis.
Collapse
Affiliation(s)
- Nour S Erekat
- Department of Anatomy, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
10
|
Red light-emitting diode irradiation regulates oxidative stress and inflammation through SPHK1/NF-κB activation in human keratinocytes. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2018; 186:31-40. [DOI: 10.1016/j.jphotobiol.2018.05.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/10/2018] [Accepted: 05/18/2018] [Indexed: 12/21/2022]
|
11
|
Zhuang J, Liu Y, Yuan Q, Liu J, Liu Y, Li H, Wang D. Blue light-induced apoptosis of human promyelocytic leukemia cells via the mitochondrial-mediated signaling pathway. Oncol Lett 2018; 15:6291-6296. [PMID: 29731847 PMCID: PMC5921239 DOI: 10.3892/ol.2018.8162] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 02/28/2018] [Indexed: 02/06/2023] Open
Abstract
Acute promyelocytic leukemia is frequently associated with dizziness, fever, nausea, hematochezia and anemia. Blue light, or light with wavelengths of 400–480 nm, transmits high levels of energy. The aim of the present study was to determine the pro-apoptotic effects of blue light (wavelength, 456 nm; radiation power, 0.25 mW/cm2) and the underlying mechanisms in a human promyelocytic leukemia cell line (HL60). Blue light reduced the viability and enhanced the mortality of HL60 cells in a time-dependent manner. Exposure to blue light for 24 h caused depolarization of the mitochondrial membrane potential and the overproduction of reactive oxygen species in HL60 cells. In a nude mouse model, 9-day exposure to blue light markedly suppressed the growth of HL60-xenografted tumors; however, it had no effect on hepatic and renal tissues. In addition, blue light abrogated the expression of B-cell lymphoma (Bcl)-2 and Bcl extra-long, while enhancing the levels of Bcl-2-associated X protein, cytochrome c, and cleaved caspases-3 and −9 in tumor tissues. The results suggested that the pro-apoptotic effects of blue light in human promyelocytic leukemia cells may be associated with the mitochondrial apoptosis signaling pathway.
Collapse
Affiliation(s)
- Jianjian Zhuang
- State Key Laboratory of Superhard Materials, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Yange Liu
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Qingxia Yuan
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Junsong Liu
- State Key Laboratory of Superhard Materials, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Yan Liu
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Hongdong Li
- State Key Laboratory of Superhard Materials, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Di Wang
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| |
Collapse
|
12
|
Agarwal A, Kasinathan A, Ganesan R, Balasubramanian A, Bhaskaran J, Suresh S, Srinivasan R, Aravind KB, Sivalingam N. Curcumin induces apoptosis and cell cycle arrest via the activation of reactive oxygen species-independent mitochondrial apoptotic pathway in Smad4 and p53 mutated colon adenocarcinoma HT29 cells. Nutr Res 2018; 51:67-81. [PMID: 29673545 DOI: 10.1016/j.nutres.2017.12.011] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 12/31/2017] [Accepted: 12/31/2017] [Indexed: 11/25/2022]
Abstract
Curcumin is a natural dietary polyphenol compound that has various pharmacological activities such as antiproliferative and cancer-preventive activities on tumor cells. Indeed, the role reactive oxygen species (ROS) generated by curcumin on cell death and cell proliferation inhibition in colon cancer is poorly understood. In the present study, we hypothesized that curcumin-induced ROS may promote apoptosis and cell cycle arrest in colon cancer. To test this hypothesis, the apoptosis-inducing potential and cell cycle inhibition effect of ROS induced by curcumin was investigated in Smd4 and p53 mutated HT-29 colon adenocarcinoma cells. We found that curcumin treatment significantly increased the level of ROS in HT-29 cells in a dose- and time-dependent manner. Furthermore, curcumin treatment markedly decreased the cell viability and proliferation potential of HT-29 cells in a dose- and time-dependent manner. Conversely, generation of ROS and inhibitory effect of curcumin on HT-29 cells were abrogated by N-acetylcysteine treatment. In addition, curcumin treatment did not show any cytotoxic effects on HT-29 cells. Furthermore, curcumin-induced ROS generation caused the DNA fragmentation, chromatin condensation, and cell nuclear shrinkage and significantly increased apoptotic cells in a dose- and time-dependent manner in HT-29 cells. However, pretreatment of N-acetylcysteine inhibited the apoptosis-triggering effect of curcumin-induced ROS in HT-29 cells. In addition, curcumin-induced ROS effectively mediated cell cycle inhibition in HT-29 cells. In conclusion, our data provide the first evidence that curcumin induces ROS independent apoptosis and cell cycle arrest in colon cancer cells that carry mutation on Smad4 and p53.
Collapse
Affiliation(s)
- Ayushi Agarwal
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamilnadu, India
| | - Akiladdevi Kasinathan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamilnadu, India
| | - Ramamoorthi Ganesan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamilnadu, India
| | - Akhila Balasubramanian
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamilnadu, India
| | - Jahnavi Bhaskaran
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamilnadu, India
| | - Samyuktha Suresh
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamilnadu, India
| | - Revanth Srinivasan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamilnadu, India
| | - K B Aravind
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamilnadu, India
| | - Nageswaran Sivalingam
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamilnadu, India.
| |
Collapse
|
13
|
Shi XJ, Yu B, Wang JW, Qi PP, Tang K, Huang X, Liu HM. Structurally novel steroidal spirooxindole by241 potently inhibits tumor growth mainly through ROS-mediated mechanisms. Sci Rep 2016; 6:31607. [PMID: 27527552 PMCID: PMC4985843 DOI: 10.1038/srep31607] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 07/21/2016] [Indexed: 12/13/2022] Open
Abstract
Cancer cells always have increased ROS levels, thus making them more vulnerable to persistent endogenous oxidative stress. The biochemical difference between cancer and normal cells could be exploited to achieve selective cancer cell killing by exogenous ROS-producing agents. Herein we described a structurally novel steroidal spirooxindole by241 and its anticancer efficacy. By241 exhibited potent inhibition against human cancer cells and less toxic to normal cells. By241 concentration-dependently induced apoptosis of MGC-803 and EC9706 cells, accompanied with the mitochondrial dysfunction and increased ROS levels. NAC can completely restore the decreased cell viability of MGC-803 cells caused by by241, suggesting ROS-mediated mechanisms. The expression levels of proteins involved in the mitochondrion-related pathways were detected, showing increased expression of proapoptotic proteins and decreased expression of anti-apoptotic proteins, and activation of caspases-9/-3, but without activating caspase-8 expression. Pretreatment with Z-VAD-FMK partially rescued by241-induced apoptosis of MGC-803 cells. Additionally, by241 inhibited mTOR, activated p53 and its downstream proteins, cleaved MDM2 and PI3K/AKT as well as NF-κB signaling pathway. In vivo experiments showed that by241 did not have significant acute oral toxicity and exerted good anticancer efficacy against MGC-803 bearing mice models. Therefore, by241 may serve as a lead for further development for cancer therapy.
Collapse
Affiliation(s)
- Xiao-Jing Shi
- School of Pharmaceutical Sciences & Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, China
| | - Bin Yu
- School of Pharmaceutical Sciences & Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, China
| | - Jun-Wei Wang
- School of Pharmaceutical Sciences & Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, China
| | - Ping-Ping Qi
- School of Pharmaceutical Sciences & Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, China
| | - Kai Tang
- School of Pharmaceutical Sciences & Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, China
| | - Xin Huang
- School of Pharmaceutical Sciences & Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, China
| | - Hong-Min Liu
- School of Pharmaceutical Sciences & Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
14
|
Rabaça AN, Arruda DC, Figueiredo CR, Massaoka MH, Farias CF, Tada DB, Maia VC, Silva Junior PI, Girola N, Real F, Mortara RA, Polonelli L, Travassos LR. AC-1001 H3 CDR peptide induces apoptosis and signs of autophagy in vitro and exhibits antimetastatic activity in a syngeneic melanoma model. FEBS Open Bio 2016; 6:885-901. [PMID: 27642552 PMCID: PMC5011487 DOI: 10.1002/2211-5463.12080] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 04/20/2016] [Accepted: 04/21/2016] [Indexed: 12/19/2022] Open
Abstract
Antibody‐derived peptides modulate functions of the immune system and are a source of anti‐infective and antitumor substances. Recent studies have shown that they comprise amino acid sequences of immunoglobulin complementarity‐determining regions, but also fragments of constant regions. VH CDR3 of murine mAb AC‐1001 displays antimetastatic activities using B16F10‐Nex2 murine melanoma cells in a syngeneic model. The peptide was cytotoxic in vitro in murine and human melanoma cells inducing reactive oxygen species (ROS) and apoptosis by the intrinsic pathway. Signs of autophagy were also suggested by the increased expression of LC3/LC3II and Beclin 1 and by ultrastructural evidence. AC‐1001 H3 bound to both G‐ and F‐actin and inhibited tumor cell migration. These results are important evidence of the antitumor activity of Ig CDR‐derived peptides.
Collapse
Affiliation(s)
- Aline N Rabaça
- Unidade de Oncologia Experimental (UNONEX) Universidade Federal de São Paulo (UNIFESP) Brazil
| | - Denise C Arruda
- Unidade de Oncologia Experimental (UNONEX) Universidade Federal de São Paulo (UNIFESP) Brazil; Núcleo Integrado de Biotecnologia Universidade de Mogi das Cruzes Brazil
| | - Carlos R Figueiredo
- Unidade de Oncologia Experimental (UNONEX) Universidade Federal de São Paulo (UNIFESP) Brazil
| | - Mariana H Massaoka
- Unidade de Oncologia Experimental (UNONEX) Universidade Federal de São Paulo (UNIFESP) Brazil
| | - Camyla F Farias
- Unidade de Oncologia Experimental (UNONEX) Universidade Federal de São Paulo (UNIFESP) Brazil
| | - Dayane B Tada
- Departamento de Ciência e Tecnologia Universidade Federal de São Paulo (UNIFESP) São José dos Campos Brazil
| | | | - Pedro I Silva Junior
- Laboratório Especial de Toxinologia Aplicada Instituto Butantan São Paulo Brazil
| | - Natalia Girola
- Unidade de Oncologia Experimental (UNONEX) Universidade Federal de São Paulo (UNIFESP) Brazil
| | - Fernando Real
- Departamento de Parasitologia Universidade Federal de São Paulo (UNIFESP) Brazil
| | - Renato A Mortara
- Departamento de Parasitologia Universidade Federal de São Paulo (UNIFESP) Brazil
| | - Luciano Polonelli
- Microbiology and Virology Unit Department of Biomedical Biotechnological and Translational Sciences Universitá degli Studi di Parma Italy
| | - Luiz R Travassos
- Unidade de Oncologia Experimental (UNONEX) Universidade Federal de São Paulo (UNIFESP) Brazil
| |
Collapse
|
15
|
Tvrdá E, Tušimová E, Kováčik A, Paál D, Libová Ľ, Lukáč N. Protective Effects of Quercetin on Selected Oxidative Biomarkers in Bovine Spermatozoa Subjected to Ferrous Ascorbate. Reprod Domest Anim 2016; 51:524-37. [DOI: 10.1111/rda.12714] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 05/16/2016] [Indexed: 12/17/2022]
Affiliation(s)
- E Tvrdá
- Department of Animal Physiology; Faculty of Biotechnology and Food Sciences; Slovak University of Agriculture in Nitra; Nitra Slovakia
| | - E Tušimová
- AgroBioTech Research Centre; Slovak University of Agriculture in Nitra; Nitra Slovakia
| | - A Kováčik
- Department of Animal Physiology; Faculty of Biotechnology and Food Sciences; Slovak University of Agriculture in Nitra; Nitra Slovakia
| | - D Paál
- Department of Botany and Genetics; Faculty of Natural Sciences; Constantine the Philosopher University in Nitra; Nitra Slovakia
| | - Ľ Libová
- St. Ladislav Institute of Health and Social Sciences; St. Elizabeth University of Health and Social Sciences; Bratislava Slovakia
| | - N Lukáč
- Department of Animal Physiology; Faculty of Biotechnology and Food Sciences; Slovak University of Agriculture in Nitra; Nitra Slovakia
| |
Collapse
|
16
|
Para-Phenylenediamine Induces Apoptotic Death of Melanoma Cells and Reduces Melanoma Tumour Growth in Mice. Biochem Res Int 2016; 2016:3137010. [PMID: 27293892 PMCID: PMC4886052 DOI: 10.1155/2016/3137010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 03/28/2016] [Accepted: 04/04/2016] [Indexed: 01/08/2023] Open
Abstract
Melanoma is one of the most aggressive forms of cancer, usually resistant to standard chemotherapeutics. Despite a huge number of clinical trials, any success to find a chemotherapeutic agent that can effectively destroy melanoma is yet to be achieved. Para-phenylenediamine (p-PD) in the hair dyes is reported to purely serve as an external dyeing agent. Very little is known about whether p-PD has any effect on the melanin producing cells. We have demonstrated p-PD mediated apoptotic death of both human and mouse melanoma cells in vitro. Mouse melanoma tumour growth was also arrested by the apoptotic activity of intraperitoneal administration of p-PD with almost no side effects. This apoptosis is shown to occur primarily via loss of mitochondrial membrane potential (MMP), generation of reactive oxygen species (ROS), and caspase 8 activation. p-PD mediated apoptosis was also confirmed by the increase in sub-G0/G1 cell number. Thus, our experimental observation suggests that p-PD can be a potential less expensive candidate to be developed as a chemotherapeutic agent for melanoma.
Collapse
|
17
|
Kashyap MK, Kumar D, Jones H, Amaya-Chanaga CI, Choi MY, Melo-Cardenas J, Ale-Ali A, Kuhne MR, Sabbatini P, Cohen LJ, Shelat SG, Rassenti LZ, Kipps TJ, Cardarelli PM, Castro JE. Ulocuplumab (BMS-936564 / MDX1338): a fully human anti-CXCR4 antibody induces cell death in chronic lymphocytic leukemia mediated through a reactive oxygen species-dependent pathway. Oncotarget 2016; 7:2809-22. [PMID: 26646452 PMCID: PMC4823073 DOI: 10.18632/oncotarget.6465] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 10/22/2015] [Indexed: 02/07/2023] Open
Abstract
The CXCR4 receptor (Chemokine C-X-C motif receptor 4) is highly expressed in different hematological malignancies including chronic lymphocytic leukemia (CLL). The CXCR4 ligand (CXCL12) stimulates CXCR4 promoting cell survival and proliferation, and may contribute to the tropism of leukemia cells towards lymphoid tissues. Therefore, strategies targeting CXCR4 may constitute an effective therapeutic approach for CLL. To address that question, we studied the effect of Ulocuplumab (BMS-936564), a fully human IgG4 anti-CXCR4 antibody, using a stroma--CLL cells co-culture model. We found that Ulocuplumab (BMS-936564) inhibited CXCL12 mediated CXCR4 activation-migration of CLL cells at nanomolar concentrations. This effect was comparable to AMD3100 (Plerixafor--Mozobil), a small molecule CXCR4 inhibitor. However, Ulocuplumab (BMS-936564) but not AMD3100 induced apoptosis in CLL at nanomolar concentrations in the presence or absence of stromal cell support. This pro-apoptotic effect was independent of CLL high-risk prognostic markers, was associated with production of reactive oxygen species and did not require caspase activation. Overall, these findings are evidence that Ulocuplumab (BMS-936564) has biological activity in CLL, highlight the relevance of the CXCR4-CXCL12 pathway as a therapeutic target in CLL, and provide biological rationale for ongoing clinical trials in CLL and other hematological malignancies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Michelle R. Kuhne
- 3 Bristol-Myers Squibb, Department of Cell Biology and Physiology, Redwood City, CA, USA
| | - Peter Sabbatini
- 4 Department of Early Clinical and Translational Research, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Lewis J. Cohen
- 4 Department of Early Clinical and Translational Research, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Suresh G. Shelat
- 4 Department of Early Clinical and Translational Research, Bristol-Myers Squibb, Princeton, NJ, USA
| | | | - Thomas J. Kipps
- 1 UCSD-Moores Cancer Center, La Jolla, CA, USA
- 2 CLL Research Consortium, La Jolla, CA, USA
| | - Pina M. Cardarelli
- 3 Bristol-Myers Squibb, Department of Cell Biology and Physiology, Redwood City, CA, USA
| | - Januario E. Castro
- 1 UCSD-Moores Cancer Center, La Jolla, CA, USA
- 2 CLL Research Consortium, La Jolla, CA, USA
| |
Collapse
|
18
|
Green Chemistry Approach for Synthesis of Effective Anticancer Palladium Nanoparticles. Molecules 2015; 20:22476-98. [PMID: 26694334 PMCID: PMC6332282 DOI: 10.3390/molecules201219860] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 12/09/2015] [Accepted: 12/10/2015] [Indexed: 12/20/2022] Open
Abstract
The purpose of this study was to design and synthesize Palladium nanoparticles (PdNPs) using an environmentally friendly approach and evaluate the in vitro efficacy of PdNPs in human ovarian cancer A2780 cells. Ultraviolet-Visible (UV-Vis) spectroscopy was used to monitor the conversion of Pd(II) ions to Pd(0)NPs. X-ray diffraction (XRD) revealed the crystallinity of the as-synthesized PdNPs and Fourier transform infrared spectroscopy (FTIR) further confirmed the role of the leaf extract of Evolvulus alsinoides as a reducing and stabilizing agent for the synthesis of PdNPs. Dynamic light scattering (DLS) and transmission electron microscopy (TEM) showed that the average size of the NPs was 5 nm. After a 24-h exposure to PdNPs, cell viability and light microscopy assays revealed the dose-dependent toxicity of the PdNPs. Furthermore, the dose-dependent cytotoxicity of the PdNPs was confirmed by lactate dehydrogenase (LDH), increased reactive oxygen species (ROS) generation, activation of PdNPs-induced autophagy, impairment of mitochondrial membrane potential (MMP), enhanced caspase-3 activity, and detection of TUNEL-positive cells. Our study demonstrates a single, simple, dependable and green approach for the synthesis of PdNPs using leaf extracts of Evolvulus alsinoides. Furthermore, the in vitro efficacy of PdNPs in human ovarian cancer cells suggests that it could be an effective therapeutic agent for cancer therapy.
Collapse
|
19
|
Balakrishnan P, Song CK, Jahn A, Cho HJ. Ceramide and N,N,N-Trimethylphytosphingosine-Iodide (TMP-I)-Based Lipid Nanoparticles for Cancer Therapy. Pharm Res 2015; 33:206-16. [PMID: 26337769 DOI: 10.1007/s11095-015-1780-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 08/13/2015] [Indexed: 01/22/2023]
Abstract
PURPOSE To evaluate the anti-tumor effect of ceramide or trimethylphytosphingosine-iodide (TMP-I) containing solid lipid nanoparticles (SLNs) prepared using trymyristin, phosphatidylcholine (PC), and Pluronic P85 (P85) for intravenous delivery of docetaxel. METHODS Docetaxel-loaded SLNs using ceramide or TMP-I at 3.22% (w/w) with a mean diameter of 89-137 nm were successfully prepared by high pressure homogenization. The prepared nanoparticles were characterized by particle size, zeta potential, drug content, and TEM analysis. Cellular uptake and cytotoxicity were studied using adriamycin-resistant breast cancer (MCF-7/ADR) cells. The optimized formulation's dissolution profile, pharmacokinetics, and antitumor effect in mice tumor model were compared with that of control (Taxotere(®)). RESULTS The drug release rate of docetaxel from SLNs was lower than that of control (Taxotere(®)). The prepared SLNs showed higher cellular uptake of docetaxel compared to that of Taxotere(®) in MCF-7/ADR cell lines, which was further confirmed by the confocal laser scanning microscopy (CLSM) study using coumarin 6 (C6). Prepared SLNs exhibited significantly increased antitumor efficacy, compared to Taxotere(®), in MCF-7/ADR cells. In vivo pharmacokinetic study in rats (at 10 mg/kg dose) showed that the SLNs significantly reduced in vivo clearance of drug than Taxotere(®). Interestingly, ceramide and TMP-I SLNs efficiently inhibited the tumor growth compared to Taxotere(®) in MCF-7/ADR tumor xenografted mouse model. CONCLUSION This work showed that TMP-I and ceramide SLNs not only significantly enhanced systemic exposure of drug, but also increased antitumor efficacy compared to Taxotere(®) and control SLN.
Collapse
Affiliation(s)
- Prabagar Balakrishnan
- College of Pharmacy, Hanyang University, Ansan, 426-791, Republic of Korea. .,School of Medicine, Stony Brook University, HSC, Stony Brook, NY, 11794, USA.
| | - Chung Kil Song
- Institute of Nano Science and Technology, Hanyang University, Seoul, 133-791, Republic of Korea
| | - Alexander Jahn
- Department of Bio-Chemical Engineering, Graduate School, Dongseo University, Busan, 617-716, Republic of Korea
| | - Hyun-Jong Cho
- College of Pharmacy, Kangwon National University, Chuncheon, 200-701, Republic of Korea.
| |
Collapse
|
20
|
Geng YD, Zhang C, Shi YM, Xia YZ, Guo C, Yang L, Kong LY. Icariside II-induced mitochondrion and lysosome mediated apoptosis is counterbalanced by an autophagic salvage response in hepatoblastoma. Cancer Lett 2015; 366:19-31. [PMID: 26118776 DOI: 10.1016/j.canlet.2015.05.032] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 05/07/2015] [Accepted: 05/21/2015] [Indexed: 01/21/2023]
Abstract
In this study, the anti-cancer effect of Icariside II (IS), a natural plant flavonoid, against hepatoblastoma cells and the underlying mechanisms were investigated. The in vitro and in vivo studies show that IS decreased the viability of human hepatoblastoma HepG2 cells in a concentration- and time-dependent manner and inhibited tumor growth in mice transplanted with H22 liver carcinomas. IS impaired mitochondria and lysosomes as evidenced by signs of induced mitochondrial and lysosomal membrane permeabilization, resulting in caspase activation and apoptosis. SQSTM1 up-regulation and autophagic flux measurements demonstrated that IS exposure also impaired autophagosome degradation which resulted in autophagosome accumulation, which plays a pro-survival role as the genetic knockdown of LC3B further sensitized the IS-treated cells. Electron microscopy images showed that autophagosome engulfs IS-impaired mitochondria and lysosomes, thus blocking cytotoxicity induced by further leakage of the hydrolases from lysosomes and pro-apoptosis members from mitochondria. In conclusion, these data suggest that IS plays multiple roles as a promising chemotherapeutic agent that induces cell apoptosis involving both mitochondrial and lysosomal damage.
Collapse
Affiliation(s)
- Ya-di Geng
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Chao Zhang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Ya-Min Shi
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Yuan-Zheng Xia
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Chao Guo
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Lei Yang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China.
| | - Ling-Yi Kong
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
21
|
Song S, Oh S, Lim KT. The proteins (12 and 15 kDa) isolated from heat-killedLactobacillus plantarumL67 induces apoptosis in HT-29 cells. Cell Biochem Funct 2015; 33:89-96. [DOI: 10.1002/cbf.3094] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Revised: 01/08/2015] [Accepted: 01/12/2015] [Indexed: 01/16/2023]
Affiliation(s)
- S. Song
- Division of Animal Science; Chonnam National University; Gwangju Korea
| | - S. Oh
- Division of Animal Science; Chonnam National University; Gwangju Korea
| | - K. T. Lim
- Division of Animal Science; Chonnam National University; Gwangju Korea
| |
Collapse
|
22
|
Song W, Hu P, Shan Y, Du M, Liu A, Ye R. Cartilage polysaccharide induces apoptosis in K562 cells through a reactive oxygen species-mediated caspase pathway. Food Funct 2014; 5:2486-93. [PMID: 25112602 DOI: 10.1039/c4fo00476k] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this study, a polysaccharide (PS) was successfully extracted from porcine cartilage and its effect on chronic myeloid leukemia was examined using human K562 cells. The results of cell proliferation assays indicated that the PS inhibited cancer cell growth at different concentrations. Morphological and biochemical changes characteristic of apoptosis were observed and confirmed by PI staining and TUNEL assay. The nuclear DNA, RNA and proteins of the cancer cells subjected to PS treatment were irreversibly destroyed by reactive oxygen species (ROS), additionally, the ROS effected on the cells directly. The apoptotic signals altered the permeability of the mitochondrial outer membrane, thereby resulted in the release of apoptotic factors into the cytoplasm that induced apoptosis. As caspase-3/7, 8 and 9 were expressed, it was speculated that both intrinsic and extrinsic pathways were involved in the PS-induced apoptosis.
Collapse
Affiliation(s)
- Wei Song
- School of Food Science and Engineering, Harbin Institute of Technology, Harbin 150090, China.
| | | | | | | | | | | |
Collapse
|
23
|
Figueiredo CR, Matsuo AL, Pereira FV, Rabaça AN, Farias CF, Girola N, Massaoka MH, Azevedo RA, Scutti JAB, Arruda DC, Silva LP, Rodrigues EG, Lago JHG, Travassos LR, Silva RMG. Pyrostegia venusta heptane extract containing saturated aliphatic hydrocarbons induces apoptosis on B16F10-Nex2 melanoma cells and displays antitumor activity in vivo. Pharmacogn Mag 2014; 10:S363-76. [PMID: 24991116 PMCID: PMC4078348 DOI: 10.4103/0973-1296.133284] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 09/25/2013] [Accepted: 05/28/2014] [Indexed: 12/30/2022] Open
Abstract
Background: Pyrostegia venusta (Ker. Gawl.) Miers (Bignoniacea) is a medicinal plant from the Brazilian Cerrado used to treat leucoderma and common diseases of the respiratory system. Objective: To investigate the antitumor activity of P.venusta extracts against melanoma. Materials and Methods: The cytotoxic activity and tumor induced cell death of heptane extract (HE) from P. venusta flowers was evaluated against murine melanoma B16F10-Nex2 cells in vitro and in a syngeneic model in vivo. Results: We found that HE induced apoptosis in melanoma cells by disruption of the mitochondrial membrane potential, induction of reactive oxygen species and late apoptosis evidenced by plasma membrane blebbing, cell shrinkage, chromatin condensation and DNA fragmentation, exposure of phosphatidylserine on the cell surface and activation of caspase-2,-3,-8,-9. HE was also protective against singeneyc subcutaneous melanoma HE compounds were also able to induce cell cycle arrest at G2/M phases on tumor cells. On fractionation of HE in silica gel we isolated a cytotoxic fraction that contained a mixture of saturated hydrocarbons identified by 1H NMR and GC-MS analyses. Predominant species were octacosane (C28H58-36%) and triacontane (C30H62-13%), which individually showed significant cytotoxic activity against murine melanoma B16F10-Nex2 cells in vitro and a very promising antitumor protection against subcutaneous melanoma in vivo. Conclusion: The results suggest that the components of the heptane extract, mainly octasane and triacontane, which showed antitumor properties in experimental melanoma upon regional administration, might also be therapeutic in human cancer, such as in the mostly epidermal and slowly invasive melanomas, such as acral lentiginous melanoma, as an adjuvant treatment to surgical excision.
Collapse
Affiliation(s)
- Carlos R Figueiredo
- Departments of Microbiology, Immunology and Parasitology, Cell Biology Division and Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Alisson L Matsuo
- Departments of Microbiology, Immunology and Parasitology, Cell Biology Division and Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Felipe V Pereira
- Departments of Microbiology, Immunology and Parasitology, Cell Biology Division and Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Aline N Rabaça
- Departments of Microbiology, Immunology and Parasitology, Cell Biology Division and Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Camyla F Farias
- Departments of Microbiology, Immunology and Parasitology, Cell Biology Division and Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Nátalia Girola
- Departments of Microbiology, Immunology and Parasitology, Cell Biology Division and Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Mariana H Massaoka
- Departments of Microbiology, Immunology and Parasitology, Cell Biology Division and Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Ricardo A Azevedo
- Departments of Microbiology, Immunology and Parasitology, Cell Biology Division and Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Jorge A B Scutti
- Departments of Microbiology, Immunology and Parasitology, Cell Biology Division and Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Denise C Arruda
- Departments of Microbiology, Immunology and Parasitology, Cell Biology Division and Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Luciana P Silva
- Department of Biological Sciences, Phytochemistry Laboratory. Universidade Estadual Paulista (UNESP), Assis, São Paulo State, Brazil
| | - Elaine G Rodrigues
- Departments of Microbiology, Immunology and Parasitology, Cell Biology Division and Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - João Henrique G Lago
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo (UNIFESP), Diadema, São Paulo, Brazil
| | - Luiz R Travassos
- Departments of Microbiology, Immunology and Parasitology, Cell Biology Division and Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Regildo M G Silva
- Department of Biological Sciences, Phytochemistry Laboratory. Universidade Estadual Paulista (UNESP), Assis, São Paulo State, Brazil
| |
Collapse
|
24
|
Zhang J, Wang Y, Li G, Yu H, Xie X. Down-regulation of nicotinamide N-methyltransferase induces apoptosis in human breast cancer cells via the mitochondria-mediated pathway. PLoS One 2014; 9:e89202. [PMID: 24558488 PMCID: PMC3928407 DOI: 10.1371/journal.pone.0089202] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 01/16/2014] [Indexed: 12/31/2022] Open
Abstract
Nicotinamide N-methyltransferase (NNMT) has been found involved in cell proliferation of several malignancies. However, the functional role of NNMT in breast cancer has not been elucidated. In the present study, we showed that NNMT was selectively expressed in some breast cancer cell lines, down-regulation of NNMT expression in Bcap-37 and MDA-MB-231 cell lines by NNMT shRNA significantly inhibited cell growth in vitro, decreased tumorigenicity in mice and induced apoptosis. The silencing reciprocal effect of NNMT was confirmed by over-expressing NNMT in the MCF-7 and SK-BR-3 breast cancer cell lines which lack constitutive expression of NNMT. In addition, down-regulation of NNMT expression resulted in reducing expression of Bcl-2 and Bcl-xL, up-regulation of Bax, Puma, cleaved caspase-9, cleaved caspase-3 and cleaved PARP, increasing reactive oxygen species production and release of cytochrome c from mitochondria, and decreasing the phosphorylation of Akt and ERK1/2. These data suggest that down-regulation of NNMT induces apoptosis via the mitochondria-mediated pathway in breast cancer cells.
Collapse
Affiliation(s)
- Jun Zhang
- Clinical Laboratory, Sir RunRun Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yanzhong Wang
- Clinical Laboratory, Sir RunRun Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Guiling Li
- Clinical Laboratory, Sir RunRun Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haitao Yu
- Clinical Laboratory, Sir RunRun Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xinyou Xie
- Clinical Laboratory, Sir RunRun Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
- * E-mail:
| |
Collapse
|
25
|
Plano D, Amin S, Sharma AK. Importance of sphingosine kinase (SphK) as a target in developing cancer therapeutics and recent developments in the synthesis of novel SphK inhibitors. J Med Chem 2014; 57:5509-24. [PMID: 24471412 DOI: 10.1021/jm4011687] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Sphingosine kinase (SphK) is an oncogenic lipid kinase that regulates the sphingolipid metabolic pathway that has been shown to play a role in numerous hyperproliferative/inflammatory diseases. The SphK isoforms (SphK1 and SphK2) catalyze the conversion of the proapoptotic substrate d-erythrosphingosine to the promitogenic/migratory product sphingosine 1-phosphate (S1P). Accumulation of S1P has been linked to the development/progression of cancer and various other diseases including, but not limited to, asthma, inflammatory bowel disease, rheumatoid arthritis, and diabetic nephropathy. SphK therefore represents a potential new target for developing novel therapeutics for cancer and other diseases. This finding has stimulated the development and evaluation of numerous SphK inhibitors over the past decade or so. In this review, we highlight the recent advancement in the field of SphK inhibitors including SphK1 and SphK2 specific inhibitors. Both sphingolipid based and nolipidic small molecule inhibitors and their importance in treatment of cancer and other diseases are discussed.
Collapse
Affiliation(s)
- Daniel Plano
- Department of Pharmacology, Penn State Hershey Cancer Institute, CH72, Penn State College of Medicine , 500 University Drive, Hershey, Pennsylvania 17033, United States
| | | | | |
Collapse
|
26
|
Zhang H, Li X, Zhang Y, Luan X. Luteolin induces apoptosis by activating Fas signaling pathway at the receptor level in laryngeal squamous cell line Hep-2 cells. Eur Arch Otorhinolaryngol 2014; 271:1653-9. [DOI: 10.1007/s00405-014-2903-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 01/17/2014] [Indexed: 11/30/2022]
|
27
|
Zhang H, Cai C, Shi C, Cao H, Han Z, Jia X. Cadmium-induced oxidative stress and apoptosis in the testes of frog Rana limnocharis. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2012; 122-123:67-74. [PMID: 22728207 DOI: 10.1016/j.aquatox.2012.05.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 05/21/2012] [Accepted: 05/29/2012] [Indexed: 06/01/2023]
Abstract
This study explored the genetic damage induced by cadmium exposure in the testes of Rana limnocharis. Healthy adult frogs were exposed to 2.5, 5, 7.5, or 10 mg/L of cadmium solution for 14 days. The results showed that exposure to these concentrations increased the levels of reactive oxygen species and malondialdehyde content in the testes, clearly indicating a dose-effect relationship. Moreover, the same dosages of Cd(2+) solution increased glutathione (reduced) content, with the values being significantly different from those observed in the control group (P<0.01). The comet assay results demonstrated that the DNA damage rate, tail length, and tail moment of samples obtained from frogs exposed to 2.5-7.5 mg/L of cadmium solution significantly increased compared with those of samples obtained from the control group (P<0.01). These findings suggest that cadmium can induce free radical generation, followed by lipid peroxidation and DNA damage. Ultrastructural observation revealed vacuoles in the spermatogenic cells, cell dispersion, incomplete cell structures, and deformed nucleoli. Moreover, cadmium exposure induced significant down-regulation of Bcl-2 expression and up-regulation of Bax and caspase-3 expressions. Taken together, these data indicate that cadmium can induce testicular cell apoptosis in R. limnocharis. Exploring the effects of cadmium on the mechanism of reproductive toxicity in amphibians will help provide a scientific basis accounting for the global population decline in amphibian species.
Collapse
Affiliation(s)
- Hangjun Zhang
- Department of Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang Province, China
| | | | | | | | | | | |
Collapse
|
28
|
PEG-liposomal oxaliplatin induces apoptosis in human colorectal cancer cells via Fas/FasL and caspase-8. Cell Biol Int 2012; 36:289-96. [PMID: 21888623 DOI: 10.1042/cbi20100825] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Since cellular uptake of PEG [poly(ethylene glycol)]-liposomal L-OHP (oxaliplatin) induces bioactive changes in CRC (colorectal cancer), we have investigated its apoptotic effect and anticancer mechanism. Human CRC SW480 cells were treated with PEG-liposomal L-OHP and a caspase-8 inhibitor [Z-IETD-FMK (benzyloxycarbonyl-Ile-Glu-Thr-dl-Asp-fluoromethylketone)]. Apoptosis was measured by FCM (flow cytometry) and TUNEL (terminal deoxynucleotidyl transferase-mediated dUTP nick-end labelling) assay. Expression of Fas/FasL and cytochrome c was detected using FCM and an immunofluorescence assay. Expression of caspase-8, Bid, caspase-9, caspase-7 and activated caspase-3 (P17) was examined by Western blot analyses. The results indicated that PEG-liposomal L-OHP (28 μg/ml L-OHP) induced marked apoptosis in SW480 cells compared with 28 μg/ml free L-OHP. The expression levels of Fas, FasL, cytochrome c, caspase-9, caspase-7 and activated caspase-3 proteins were up-regulated, with a corresponding increase in apoptosis; however, expression of caspase-8 and Bid were down-regulated as apoptosis increased. When cells were treated with Z-IETD-FMK, apoptosis was inhibited, but there was little impact on the expression of Fas, FasL, cytochrome c, Bid, caspase-9, caspase-7 and activated caspase-3. These findings indicate that PEG-liposomal L-OHP enhances the anticancer potency of the chemotherapeutic agent; moreover, Fas/FasL and caspase-8 signalling pathways play a key role in mediating PEG-liposomal L-OHP-induced apoptosis.
Collapse
|
29
|
Hoffman L, Hardej D. Ethylene bisdithiocarbamate pesticides cause cytotoxicity in transformed and normal human colon cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2012; 34:556-573. [PMID: 22824503 DOI: 10.1016/j.etap.2012.06.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 06/25/2012] [Indexed: 06/01/2023]
Abstract
The effects of the fungicides Maneb, Mancozeb, and Zineb were investigated in transformed colon cells, HT-29, Caco2 and non-transformed cells, CCD-18Co. Significant decreases in viability were observed with Maneb and Mancozeb in HT-29 and CCD-18Co (80-260μM), and Caco2 cells (40-180μM). No significant decreases in viability were observed in all cell types up to 800μM with Zineb. MnCl(2) and ZnCl(2) exposure produced no loss of viability in all cell types up to 400μM. Light microscopy confirmed viability analysis. Lipid peroxidation was observed with Maneb and Mancozeb in cell types tested (60-200μM). Caspase 3/7, 8, and 9 activities were observed with Maneb and Mancozeb in cell types tested (40-200μM). Maneb and Mancozeb treated HT-29 and Caco2 cells demonstrated increases in manganese and zinc concentrations (20-200μM). The lack of toxicity observed with Zineb, MnCl(2), and ZnCl(2) suggests that both the metal moiety and the organic portion of these fungicides together contribute to toxicity.
Collapse
Affiliation(s)
- Lisa Hoffman
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, St. John's University, Jamaica, NY 11439, USA
| | - Diane Hardej
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, St. John's University, Jamaica, NY 11439, USA.
| |
Collapse
|
30
|
The control of the balance between ceramide and sphingosine-1-phosphate by sphingosine kinase: Oxidative stress and the seesaw of cell survival and death. Comp Biochem Physiol B Biochem Mol Biol 2012; 163:26-36. [DOI: 10.1016/j.cbpb.2012.05.006] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 05/09/2012] [Accepted: 05/12/2012] [Indexed: 12/19/2022]
|
31
|
Ji Y, Ji C, Yue L, Xu H. Saponins isolated from Asparagus induce apoptosis in human hepatoma cell line HepG2 through a mitochondrial-mediated pathway. ACTA ACUST UNITED AC 2012; 19:eS1-9. [PMID: 22876162 DOI: 10.3747/co.19.1139] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Many scientific studies have shown that Asparagus officinalis has an antitumour effect and enhances human immunity, but the active components and the antitumour mechanisms are unclear. We investigated the effects of saponins isolated from Asparagus on proliferation and apoptosis in the human hepatoma cell line HepG2. METHODS HepG2 cells were treated with varying concentrations of Asparagus saponins at various times. Using mtt and flow cytometry assays, we evaluated the effects of Asparagus saponins on the growth and apoptosis of HepG2 cells. Transmission electron microscopy was used to observe the morphology of cell apoptosis. Confocal laser scanning microscopy was used to analyze intracellular calcium ion concentration, mitochondrial permeability transition pore (mptp), and mitochondrial membrane potential (mmp). Spectrophotometry was applied to quantify the activity of caspase-9 and caspase-3. Flow cytometry was used to investigate the levels of reactive oxygen species (ros) and pH, and the expressions of Bcl2, Bax, CytC, and caspase-3, in HepG2 cells. RESULTS Asparagus saponins inhibited the growth of HepG2 cells in a dose-dependent manner. The median inhibitory concentration (IC(50)) was 101.15 mg/L at 72 hours. The apoptosis morphology at 72 hours of treatment was obvious, showing cell protuberance, concentrated cytoplasm, and apoptotic bodies. The apoptotic rates at 72 hours were 30.9%, 51.7%, and 62.1% (for saponin concentrations of 50 mg/L, 100 mg/L, 200 mg/L). Treatment with Asparagus saponins for 24 hours increased the intracellular level of ros and Ca(2+), lowered the pH, activated intracellular mptp, and decreased mmp in a dose-dependent manner. Treatment also increased the activity of caspase-9 and caspase-3, downregulated the expression of Bcl2, upregulated the expression of Bax, and induced release of CytC and activation of caspase-3. CONCLUSIONS Asparagus saponins induce apoptosis in HepG2 cells through a mitochondrial-mediated and caspase-dependent pathway, suggesting that they may be a potent agent for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Y Ji
- Engineering Research Center of Natural Anticancer Drugs, Ministry of Education, Harbin, PR China
| | | | | | | |
Collapse
|
32
|
Ji YB, Ji CF, Zhang H. Laminarin induces apoptosis of human colon cancer LOVO cells through a mitochondrial pathway. Molecules 2012; 17:9947-60. [PMID: 22907156 PMCID: PMC6268208 DOI: 10.3390/molecules17089947] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Revised: 08/11/2012] [Accepted: 08/14/2012] [Indexed: 12/11/2022] Open
Abstract
Many scientific studies have shown that laminarin has anti-tumor effects, but the anti-tumor mechanism was unclear. The purpose of this study was to investigate the effect of laminarin on the induction of apoptosis in human colon cancer LOVO cells and the molecular mechanism involved. LOVO cells were treated with different concentrations of laminarin at different times. Morphology observations were performed to determine the effects of laminarin on apoptosis of LOVO cells. Flow cytometry (FCM) was used to detect the level of intracellular reactive oxygen species (ROS) and pH. Laser scanning confocal microscope (LSCM) was used to analyze intracellular calcium ion concentration, mitochondrion permeability transition pore (MPTP) and mitochondrial membrane potential (MMP). Western blotd were performed to analyze the expressions of Cyt-C, Caspase-9 and -3. The results showed the apoptosis morphology, which showed cell protuberance, concentrated cytoplasm and apoptotic bodies, was obvious after 72 h treatment. Laminarin treatment for 24 h increased the intracellular level of ROS and Ca2+; decreased pH value; activated intracellular MPTP and decreased MMP in dose-dependent manners. It also induced the release of Cyt-C and the activation of Caspase-9 and -3. In conclusion, laminarin induces LOVO cell apoptosis through a mitochondrial pathway, suggesting that it could be a potent agent for cancer prevention and treatment.
Collapse
Affiliation(s)
- Yu Bin Ji
- Engineering Research Center of Natural Anticancer Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China; E-Mail:
| | - Chen Feng Ji
- Center of Research on Life Science and Environmental Science, Harbin University of Commerce, Harbin 150076, China; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +86-451-8484-4242; Fax: +86-451-8484-4417
| | - He Zhang
- Center of Research on Life Science and Environmental Science, Harbin University of Commerce, Harbin 150076, China; E-Mail:
| |
Collapse
|
33
|
Bu TL, Jia YD, Lin JX, Mi YL, Zhang CQ. Alleviative effect of quercetin on germ cells intoxicated by 3-methyl-4-nitrophenol from diesel exhaust particles. J Zhejiang Univ Sci B 2012; 13:318-26. [PMID: 22467373 DOI: 10.1631/jzus.b1100318] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
As a component of diesel exhaust particles, 3-methyl-4-nitrophenol (4-nitro-m-cresol, PNMC) is also a metabolite of the insecticide fenitrothion and imposes hazardous effects on human health. In the present study, the alleviative effect of a common antioxidant flavonoid quercetin on mouse germ cells intoxicated by PNMC was investigated. Results showed that a single intraperitoneal injection of PNMC at 100 mg/kg induced severe testicular damage after one week. PNMC-treated mice showed a significant loss of germ cells (approximate 40% loss of round germ cells). PNMC caused an increase of hydroxyl radical and hydrogen peroxide production and lipid peroxidation, as well as a decrease in glutathione level, superoxide dismutase and glutathione peroxidase activities. Furthermore, treatment of PNMC increased expression of the pro-apoptotic protein Bax and decreased expression of the anti-apoptotic protein Bcl-XL in germ cells. In addition, testicular caspase-3 activity was significantly up-regulated and germ cell apoptosis was significantly increased in the PNMC-treated mice. In contrast, combined administration of quercetin at 75 mg/kg significantly attenuated PNMC-induced testicular toxicity. These results indicate that the antioxidant quercetin displays a remarkable protective effect on PNMC-induced oxidative damage in mouse testes and may represent an efficient supplement to attenuate reproductive toxicity by environmental toxicants to ensure healthy sperm production.
Collapse
Affiliation(s)
- Tong-liang Bu
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | | | | | | | | |
Collapse
|
34
|
Detailed analysis of apoptosis and delayed luminescence of human leukemia Jurkat T cells after proton irradiation and treatments with oxidant agents and flavonoids. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2012; 2012:498914. [PMID: 22829956 PMCID: PMC3397210 DOI: 10.1155/2012/498914] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 05/14/2012] [Indexed: 11/24/2022]
Abstract
Following previous work, we investigated in more detail the relationship between apoptosis and delayed luminescence (DL) in human leukemia Jurkat T cells under a wide variety of treatments. We used menadione and hydrogen peroxide to induce oxidative stress and two flavonoids, quercetin, and epigallocatechin gallate, applied alone or in combination with menadione or H2O2. 62 MeV proton beams were used to irradiate cells under a uniform dose of 2 or 10 Gy, respectively. We assessed apoptosis, cell cycle distributions, and DL. Menadione, H2O2 and quercetin were potent inducers of apoptosis and DL inhibitors. Quercetin decreased clonogenic survival and the NAD(P)H level in a dose-dependent manner. Proton irradiation with 2 Gy but not 10 Gy increased the apoptotic rate. However, both doses induced a substantial G2/M arrest. Quercetin reduced apoptosis and prolonged the G2/M arrest induced by radiation. DL spectroscopy indicated that proton irradiation disrupted the electron flow within Complex I of the mitochondrial respiratory chain, thus explaining the massive necrosis induced by 10 Gy of protons and also suggested an equivalent action of menadione and quercetin at the level of the Fe/S center N2, which may be mediated by their binding to a common site within Complex I, probably the rotenone-binding site.
Collapse
|
35
|
Song CK, Lee JH, Jahn A, Choi MJ, Namgoong SK, Hong SS, Chong S, Shim CK, Chung SJ, Kim DD. In vitro and in vivo evaluation of N,N,N-trimethylphytosphingosine-iodide (TMP) in liposomes for the treatment of angiogenesis and metastasis. Int J Pharm 2012; 434:191-8. [PMID: 22643227 DOI: 10.1016/j.ijpharm.2012.05.042] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 05/14/2012] [Accepted: 05/19/2012] [Indexed: 10/28/2022]
Abstract
Phytosphingosine and methyl derivatives are important mediators on cellular processes, and are associated with cell growth and death. The antitumor activity of N,N,N-trimethylphytosphingosine-iodide (TMP) as a novel potent inhibitor of angiogenesis and metastasis was evaluated in B16F10 murine melanoma cells. The results indicated that TMP itself effectively inhibited in vitro cell migration, tube formation, and the expression of angiogenic factors as well as in vivo lung metastasis. However, TMP slightly suppressed in vivo experimental tumor metastasis in its free form and induced side effects including hemolysis and local side effects. Therefore, in an attempt to reduce the toxicity and the undesirable side effects of TMP, a liposomal formulation was prepared and tested for its effectiveness. TMP liposomes retained the effectiveness of TMP in vitro while side effects were reduced, and both in vivo experimental and spontaneous tumor metastasis were significantly suppressed. These results support the conclusion that TMP effectively inhibits in vitro angiogenesis as well as in vivo metastasis, and a liposomal formulation is more efficient delivery system for TMP treatment than solution.
Collapse
Affiliation(s)
- Chung Kil Song
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Mukherjee SP, Byrne HJ. Polyamidoamine dendrimer nanoparticle cytotoxicity, oxidative stress, caspase activation and inflammatory response: experimental observation and numerical simulation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2012; 9:202-11. [PMID: 22633897 DOI: 10.1016/j.nano.2012.05.002] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 04/02/2012] [Accepted: 05/07/2012] [Indexed: 02/01/2023]
Abstract
UNLABELLED Mechanisms underlying the in vitro cytotoxicity of Polyamidoamine nano-dendrimers in human keratinocytes are explored. Previous studies demonstrated a systematic, dendrimer-generation-dependent cytotoxicity, oxidative stress, and genotoxicity. The emerging picture is of dendrimer endocytosis, endosomal rupture and subsequent mitochondrial attack and cell death. To understand the underlying mechanisms, the evolution of reactive oxygen species, intracellular glutathione, caspase activation, mitochondrial membrane potential decay, and inflammatory responses have been examined. Early-stage responses are associated with endosomal encapsulation, later-stage with mitochondrial attack. In all cases, the magnitude and evolution of responses depend on dendrimer generation and dose. The early-stage response is modelled using a rate equation approach, qualitatively reproducing the time, dose and generation dependences, using only two variable parameters. The dependence of the response on the nanoparticle physicochemical properties can thus be separated from internal cellular parameters, and responses can be quantified in terms of rate constants rather than commonly employed effective concentrations. FROM THE CLINICAL EDITOR This contribution reports on the intracellular mechanism of PAMAM dendrimer cytotoxicity in human keratinocytes. In all cases, the magnitude and evolution of responses depend on dendrimer generation and dose. Experimental data were supported by numerical simulation using only two variables. It is suggested that responses can be quantified in terms of rate constants rather than effective concentrations.
Collapse
Affiliation(s)
- Sourav Prasanna Mukherjee
- Centre for Radiation and Environmental Science (RESC), Focas Research Institute, Dublin Institute of Technology, Dublin, Ireland.
| | | |
Collapse
|
37
|
Stevenson CE, Takabe K, Nagahashi M, Milstien S, Spiegel S. Targeting sphingosine-1-phosphate in hematologic malignancies. Anticancer Agents Med Chem 2012; 11:794-8. [PMID: 21707492 DOI: 10.2174/187152011797655122] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Revised: 05/05/2011] [Accepted: 05/06/2011] [Indexed: 12/26/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a pleiotropic bioactive lipid mediator that regulates several processes important for hematologic cancer progression. S1P is generated by two sphingosine kinases, SphK1 and SphK2, and is exported outside the cell, where it activates specific cell surface S1P G-protein coupled receptors in autocrine/paracrine manner, coined "inside-out signaling". In this review, we highlight the importance of SphK1 and inside-out signaling by S1P in hematologic malignancy. We also summarize the results of studies targeting the SphK1/S1P/S1P receptor axis and the effects of the S1P receptor modulator, FTY720, in hematologic malignancy.
Collapse
Affiliation(s)
- Christina E Stevenson
- Division of Surgical Oncology, Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, 23298-0614, USA
| | | | | | | | | |
Collapse
|
38
|
Arruda DC, Santos LCP, Melo FM, Pereira FV, Figueiredo CR, Matsuo AL, Mortara RA, Juliano MA, Rodrigues EG, Dobroff AS, Polonelli L, Travassos LR. β-Actin-binding complementarity-determining region 2 of variable heavy chain from monoclonal antibody C7 induces apoptosis in several human tumor cells and is protective against metastatic melanoma. J Biol Chem 2012; 287:14912-22. [PMID: 22334655 DOI: 10.1074/jbc.m111.322362] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Complementarity-determining regions (CDRs) from monoclonal antibodies tested as synthetic peptides display anti-infective and antitumor activities, independent of the specificity of the native antibody. Previously, we have shown that the synthetic peptide C7H2, based on the heavy chain CDR 2 from monoclonal antibody C7, a mAb directed to a mannoprotein of Candida albicans, significantly reduced B16F10 melanoma growth and lung colony formation by triggering tumor apoptosis. The mechanism, however, by which C7H2 induced apoptosis in tumor cells remained unknown. Here, we demonstrate that C7H2 interacts with components of the tumor cells cytoskeleton, being rapidly internalized after binding to the tumor cell surface. Mass spectrometry analysis and in vitro validation revealed that β-actin is the receptor of C7H2 in the tumor cells. C7H2 induces β-actin polymerization and F-actin stabilization, linked with abundant generation of superoxide anions and apoptosis. Major phenotypes following peptide binding were chromatin condensation, DNA fragmentation, annexin V binding, lamin disruption, caspase 8 and 3 activation, and organelle alterations. Finally, we evaluated the cytotoxic efficacy of C7H2 in a panel of human tumor cell lines. All tumor cell lines studied were equally susceptible to C7H2 in vitro. The C7H2 amide without further derivatization significantly reduced lung metastasis of mice endovenously challenged with B16F10-Nex2 melanoma cells. No significant cytotoxicity was observed toward nontumorigenic cell lines on short incubation in vitro or in naïve mice injected with a high dose of the peptide. We believe that C7H2 is a promising peptide to be developed as an anticancer drug.
Collapse
Affiliation(s)
- Denise C Arruda
- Experimental Oncology Unit (UNONEX), Universidade Federal de São Paulo (UNIFESP), São Paulo SP 04023-062, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Liang CH, Wang GH, Chou TH, Wang SH, Lin RJ, Chan LP, So EC, Sheu JH. 5-epi-Sinuleptolide induces cell cycle arrest and apoptosis through tumor necrosis factor/mitochondria-mediated caspase signaling pathway in human skin cancer cells. Biochim Biophys Acta Gen Subj 2012; 1820:1149-57. [PMID: 22348919 DOI: 10.1016/j.bbagen.2012.02.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 02/01/2012] [Accepted: 02/04/2012] [Indexed: 12/21/2022]
Abstract
BACKGROUND Skin cancers are reportedly increasing worldwide. Developing novel anti-skin cancer drugs with minimal side effects is necessary to address this public health issue. Sinuleptolide has been demonstrated to possess anti-cancer cell activities; however, the mechanisms underlying the anti-skin cancer effects of 5-epi-sinuleptolide and sinuleptolide remain poorly understood. METHODS Apoptosis cell, cell-cycle-related regulatory factors, and mitochondria- and death receptor-dependent caspase pathway in 5-epi-sinuleptolide-induced cell apoptosis were examined using SCC25 cells. RESULTS 5-epi-Sinuleptolide inhibited human skin cancer cell growth more than did sinuleptolide. Treatment of SCC25 cells with 5-epi-sinuleptolide increased apoptotic body formation, and induced cell-cycle arrest during the G2/M phase. Notably, 5-epi-sinuleptolide up-regulated p53 and p21 expression and inhibited G2/M phase regulators of cyclin B1 and cyclin-dependent kinease 1 (CDK1) in SCC25 cells. Additionally, 5-epi-sinuleptolide induced apoptosis by mitochondria-mediated cytochrome c and Bax up-expression, down-regulated Bcl-2, and activated caspase-9 and -3. 5-epi-Sinuleptolide also up-regulated tBid, which is associated with up-regulation of tumor necrosis factor-α (TNF-α) and Fas ligand (FasL) and their cognate receptors (i.e., TNF-RI, TNF-R2 and Fas), downstream adaptor TNF-R1-associated death domain (TRADD) and Fas-associated death domain (FADD), and activated caspase-8 in SCC25 cells. CONCLUSIONS The analytical results indicate that the death receptor- and mitochondria-mediated caspase pathway is critical in 5-epi-sinuleptolide-induced apoptosis of skin cancer cells. GENERAL SIGNIFICANCE This is the first report suggesting that the apoptosis mediates the anti-tumor effect of 5-epi-sinuleptolide. The results of this study might provide useful suggestions for designing of anti-tumor drugs for skin cancer patients.
Collapse
Affiliation(s)
- Chia-Hua Liang
- Department of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan, Taiwan.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Kuno T, Tsukamoto T, Hara A, Tanaka T. Cancer chemoprevention through the induction of apoptosis by natural compounds. ACTA ACUST UNITED AC 2012. [DOI: 10.4236/jbpc.2012.32018] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
41
|
Lin ML, Lu YC, Su HL, Lin HT, Lee CC, Kang SE, Lai TC, Chung JG, Chen SS. Destabilization of CARP mRNAs by aloe-emodin contributes to caspase-8-mediated p53-independent apoptosis of human carcinoma cells. J Cell Biochem 2011; 112:1176-91. [PMID: 21308745 DOI: 10.1002/jcb.23031] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Using short hairpin RNA against p53, transient ectopic expression of wild-type p53 or mutant p53 (R248W or R175H), and a p53- and p21-dependent luciferase reporter assay, we demonstrated that growth arrest and apoptosis of FaDu (human pharyngeal squamous cell carcinoma), Hep3B (hepatoma), and MG-63 (osteosarcoma) cells induced by aloe-emodin (AE) are p53-independent. Co-immunoprecipitation and small interfering RNA (siRNA) studies demonstrated that AE caused S-phase cell cycle arrest by inducing the formation of cyclin A-Cdk2-p21 complexes through extracellular signal-regulated kinase (ERK) activation. Ectopic expression of Bcl-X(L) and siRNA-mediated Bax attenuation significantly inhibited apoptosis induced by AE. Cyclosporin A or the caspase-8 inhibitor Z-IETD-FMK blocked AE-induced loss of mitochondrial membrane potential and prevented increases in reactive oxygen species and Ca(++). Z-IETD-FMK inhibited AE-induced apoptosis, Bax expression, Bid cleavage, translocation of tBid to mitochondria, ERK phosphorylation, caspase-9 activation, and the release of cytochrome c, apoptosis-inducing factor (AIF), and endonuclease G from mitochondria. The stability of the mRNAs encoding caspase-8 and -10-associated RING proteins (CARPs) 1 and 2 was affected by AE, whereas CARP1 or 2 overexpression inhibited caspase-8 activation and apoptosis induced by AE. Collectively, our data indicate AE induces caspase-8-mediated activation of mitochondrial death pathways by decreasing the stability of CARP mRNAs in a p53-independent manner.
Collapse
Affiliation(s)
- Meng-Liang Lin
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Bu T, Mi Y, Zeng W, Zhang C. Protective Effect of Quercetin on Cadmium-Induced Oxidative Toxicity on Germ Cells in Male Mice. Anat Rec (Hoboken) 2010; 294:520-6. [DOI: 10.1002/ar.21317] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Accepted: 10/12/2010] [Indexed: 11/06/2022]
|
43
|
Jung B, Baek CY, Yang JY, Park JH, Kim JD. Anticancer therapeutic self-aggregates of sphingolipid metabolite-grafted poly(amino acid)-derivative and their enhanced intracellular delivery. J IND ENG CHEM 2010. [DOI: 10.1016/j.jiec.2010.09.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
44
|
Lee J, Lim KT. Apoptotic activity of ethanol extract from Styrax Japonica Siebold et al Zuccarini in HepG2 cells. JOURNAL OF ETHNOPHARMACOLOGY 2010; 131:210-215. [PMID: 20600761 DOI: 10.1016/j.jep.2010.06.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Revised: 06/13/2010] [Accepted: 06/14/2010] [Indexed: 05/29/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Styrax japonica Siebold et al Zuccarini (SJSZ) has been used to heal inflammation and bronchitis as an herbal plant in Korea. AIMS OF THE STUDY The purpose of the present study is to determine whether the ethanol (EtOH) extract of SJSZ induces the programmed cell death (apoptosis) in human hepatoma HepG2 cells. MATERIALS AND METHODS It was evaluated cytotoxicity using MTT assay, amount of intracellular reactive oxygen species (iROS) and Ca(2+) using fluorescence. Activities of apoptotic relevant factors [Bid, cytochrome c, caspase-9, -3, and poly-(ADP-ribose) polymerase (PARP)] were measured by Western blot. RESULTS The results in this study indicated that ethanol extract of SJSZ (75 microg/ml) stimulates to increase amount of iROS, Ca(2+), and the apoptotic relevant factors [Bid, cytochrome c, caspase-9, -3, and poly-(ADP-ribose) polymerase (PARP) in the HepG2 cells. CONCLUSION The results in this study indicated that ethanol extract of SJSZ (75 microg/ml) induces programmed cell death (apoptosis) in the HepG2 cells. Therefore, we speculate that ethanol extract of SJSZ could be used for healing of hepatocarcinoma as one of chemotherapeutic agents.
Collapse
Affiliation(s)
- Jin Lee
- Molecular Biochemistry Laboratory, Biotechnology Research Institute & Center for the Control of Animal Hazards Using Biotechnology (BK21), Chonnam National University, 300 Yongbong-Dong, Gwang-ju 500-757, South Korea
| | | |
Collapse
|
45
|
Baran I, Ganea C, Scordino A, Musumeci F, Barresi V, Tudisco S, Privitera S, Grasso R, Condorelli DF, Ursu I, Baran V, Katona E, Mocanu MM, Gulino M, Ungureanu R, Surcel M, Ursaciuc C. Effects of Menadione, Hydrogen Peroxide, and Quercetin on Apoptosis and Delayed Luminescence of Human Leukemia Jurkat T-Cells. Cell Biochem Biophys 2010; 58:169-79. [DOI: 10.1007/s12013-010-9104-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
46
|
N,N-Dimethyl phytosphingosine sensitizes HL-60/MX2, a multidrug-resistant variant of HL-60 cells, to doxorubicin-induced cytotoxicity through ROS-mediated release of cytochrome c and AIF. Apoptosis 2010; 15:982-93. [DOI: 10.1007/s10495-010-0512-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
47
|
Ekiz HA, Baran Y. Therapeutic applications of bioactive sphingolipids in hematological malignancies. Int J Cancer 2010; 127:1497-506. [DOI: 10.1002/ijc.25478] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
48
|
Lin ML, Lu YC, Chung JG, Li YC, Wang SG, NG SH, Wu CY, Su HL, Chen SS. Aloe-emodin induces apoptosis of human nasopharyngeal carcinoma cells via caspase-8-mediated activation of the mitochondrial death pathway. Cancer Lett 2010; 291:46-58. [DOI: 10.1016/j.canlet.2009.09.016] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2009] [Revised: 09/28/2009] [Accepted: 09/29/2009] [Indexed: 12/11/2022]
|
49
|
Liu CM, Zheng YL, Lu J, Zhang ZF, Fan SH, Wu DM, Ma JQ. Quercetin protects rat liver against lead-induced oxidative stress and apoptosis. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2010; 29:158-166. [PMID: 21787598 DOI: 10.1016/j.etap.2009.12.006] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Revised: 12/22/2009] [Accepted: 12/23/2009] [Indexed: 05/31/2023]
Abstract
Quercetin, a flavonoid, effectively improved the lead-induced histology changes including structure damage and leukocyte infiltration in rat liver. The present study was designed to explore the protective mechanism of quercetin against lead-induced hepatic injury. We found that quercetin markedly decreased the MDA and H(2)O(2) levels and lowered the GSH/GSSG ratio in the liver of lead-treated rat. Moreover, quercetin markedly restored Cu/Zn-SOD, Mn-SOD, CAT and GPx activities and upregulated mRNA expression levels of these proteins in the liver of lead-treated rat. Western blot analysis showed that quercetin significantly inhibited apoptosis by modulating the ratio of Bax to Bcl-2 expression and suppressing the expression of phosphorylated JNK1/2 and cleaved caspase-3 in the liver of lead-treated rat. In conclusion, these data suggest that quercetin protects the rat liver from lead-induced injury by attenuating lipid peroxidation, renewing the activities of antioxidant enzymes and inhibiting apoptosis.
Collapse
Affiliation(s)
- Chan-Min Liu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Xuzhou Normal University, No. 101, Shanghai Road, Tangshan New Area, Xuzhou City 221116, Jiangsu, PR China
| | | | | | | | | | | | | |
Collapse
|
50
|
Kim HJ, Kang JS, Park HR, Hwang YI. Neuroprotective Effects of Methanolic Extracts from Peanut Sprouts. ACTA ACUST UNITED AC 2010. [DOI: 10.5352/jls.2010.20.2.253] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|