1
|
Chen B, Zhang W, Lin C, Zhang L. A Comprehensive Review on Beneficial Effects of Catechins on Secondary Mitochondrial Diseases. Int J Mol Sci 2022; 23:ijms231911569. [PMID: 36232871 PMCID: PMC9569714 DOI: 10.3390/ijms231911569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/13/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Mitochondria are the main sites for oxidative phosphorylation and synthesis of adenosine triphosphate in cells, and are known as cellular power factories. The phrase "secondary mitochondrial diseases" essentially refers to any abnormal mitochondrial function other than primary mitochondrial diseases, i.e., the process caused by the genes encoding the electron transport chain (ETC) proteins directly or impacting the production of the machinery needed for ETC. Mitochondrial diseases can cause adenosine triphosphate (ATP) synthesis disorder, an increase in oxygen free radicals, and intracellular redox imbalance. It can also induce apoptosis and, eventually, multi-system damage, which leads to neurodegenerative disease. The catechin compounds rich in tea have attracted much attention due to their effective antioxidant activity. Catechins, especially acetylated catechins such as epicatechin gallate (ECG) and epigallocatechin gallate (EGCG), are able to protect mitochondria from reactive oxygen species. This review focuses on the role of catechins in regulating cell homeostasis, in which catechins act as a free radical scavenger and metal ion chelator, their protective mechanism on mitochondria, and the protective effect of catechins on mitochondrial deoxyribonucleic acid (DNA). This review highlights catechins and their effects on mitochondrial functional metabolic networks: regulating mitochondrial function and biogenesis, improving insulin resistance, regulating intracellular calcium homeostasis, and regulating epigenetic processes. Finally, the indirect beneficial effects of catechins on mitochondrial diseases are also illustrated by the warburg and the apoptosis effect. Some possible mechanisms are shown graphically. In addition, the bioavailability of catechins and peracetylated-catechins, free radical scavenging activity, mitochondrial activation ability of the high-molecular-weight polyphenol, and the mitochondrial activation factor were also discussed.
Collapse
|
2
|
Rinaldi DE, Ontiveros MQ, Saffioti NA, Vigil MA, Mangialavori IC, Rossi RC, Rossi JP, Espelt MV, Ferreira-Gomes MS. Epigallocatechin 3-gallate inhibits the plasma membrane Ca 2+-ATPase: effects on calcium homeostasis. Heliyon 2021; 7:e06337. [PMID: 33681501 PMCID: PMC7930289 DOI: 10.1016/j.heliyon.2021.e06337] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/16/2021] [Accepted: 02/17/2021] [Indexed: 12/27/2022] Open
Abstract
Flavonoids are natural compounds responsible for the health benefits of green tea. Some of the flavonoids present in green tea are catechins, among which are: epigallocatechin, epicatechin-3-gallate, epicatechin, catechin and epigallocatechin-3-gallate (EGCG). The latter was found to induce apoptosis, reduce reactive oxygen species, in some conditions though in others it acts as an oxidizing agent, induce cell cycle arrest, and inhibit carcinogenesis. EGCG also was found to be involved in calcium (Ca2+) homeostasis in excitable and in non-excitable cells. In this study, we investigate the effect of catechins on plasma membrane Ca2+-ATPase (PMCA), which is one of the main mechanisms that extrude Ca2+ out of the cell. Our studies comprised experiments on the isolated PMCA and on cells overexpressing the pump. Among catechins that inhibited PMCA activity, the most potent inhibitor was EGCG. EGCG inhibited PMCA activity in a reversible way favoring E1P conformation. EGCG inhibition also occurred in the presence of calmodulin, the main pump activator. Finally, the effect of EGCG on PMCA activity was studied in human embryonic kidney cells (HEK293T) that transiently overexpress hPMCA4. Results show that EGCG inhibited PMCA activity in HEK293T cells, suggesting that the effects observed on isolated PMCA occur in living cells.
Collapse
Affiliation(s)
| | | | - Nicolas A. Saffioti
- IQUIFIB – Instituto de Química y Fisicoquímica Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CONICET, Junín 956, 1113 Buenos Aires, Argentina
| | - Maximiliano A. Vigil
- IQUIFIB – Instituto de Química y Fisicoquímica Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CONICET, Junín 956, 1113 Buenos Aires, Argentina
| | - Irene C. Mangialavori
- IQUIFIB – Instituto de Química y Fisicoquímica Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CONICET, Junín 956, 1113 Buenos Aires, Argentina
| | - Rolando C. Rossi
- IQUIFIB – Instituto de Química y Fisicoquímica Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CONICET, Junín 956, 1113 Buenos Aires, Argentina
| | - Juan P. Rossi
- IQUIFIB – Instituto de Química y Fisicoquímica Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CONICET, Junín 956, 1113 Buenos Aires, Argentina
| | - María V. Espelt
- IQUIFIB – Instituto de Química y Fisicoquímica Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CONICET, Junín 956, 1113 Buenos Aires, Argentina
| | - Mariela S. Ferreira-Gomes
- IQUIFIB – Instituto de Química y Fisicoquímica Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CONICET, Junín 956, 1113 Buenos Aires, Argentina
| |
Collapse
|
3
|
Braicu C, Zanoaga O, Zimta AA, Tigu AB, Kilpatrick KL, Bishayee A, Nabavi SM, Berindan-Neagoe I. Natural compounds modulate the crosstalk between apoptosis- and autophagy-regulated signaling pathways: Controlling the uncontrolled expansion of tumor cells. Semin Cancer Biol 2020; 80:218-236. [PMID: 32502598 DOI: 10.1016/j.semcancer.2020.05.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/22/2020] [Accepted: 05/24/2020] [Indexed: 02/07/2023]
Abstract
Due to the high number of annual cancer-related deaths, and the economic burden that this malignancy affects today's society, the study of compounds isolated from natural sources should be encouraged. Most cancers are the result of a combined effect of lifestyle, environmental factors, and genetic and hereditary components. Recent literature reveals an increase in the interest for the study of phytochemicals from traditional medicine, this being a valuable resource for modern medicine to identify novel bioactive agents with potential medicinal applications. Phytochemicals are components of traditional medicine that are showing promising application in modern medicine due to their antitumor activities. Recent studies regarding two major mechanisms underlying cancer development and regulation, apoptosis and autophagy, have shown that the signaling pathways of both these processes are significantly interconnected through various mechanisms of crosstalk. Phytochemicals are able to activate pro-autophagic and pro-apoptosis mechanisms. Understanding the molecular mechanism involved in apoptosis-autophagy relationship modulated by phytochemicals plays a key role in development of a new therapeutic strategy for cancer treatment. The purpose of this review is to outline the bioactive properties of the natural phytochemicals with validated antitumor activity, focusing particularly on their role in the regulation of apoptosis and autophagy crosstalk that triggers the uncontrolled expansion of tumor cells. Furthermore, we have also critically discussed the limitations and challenges of existing research strategies and the prospective research directions in this field.
Collapse
Affiliation(s)
- Cornelia Braicu
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 40015, Cluj-Napoca, Romania
| | - Oana Zanoaga
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 40015, Cluj-Napoca, Romania
| | - Alina-Andreea Zimta
- MEDFUTURE-Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 40015, Cluj-Napoca, Romania
| | - Adrian Bogdan Tigu
- MEDFUTURE-Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 40015, Cluj-Napoca, Romania; Babeș-Bolyai University, Faculty of Biology and Geology, 42 Republicii Street, 400015, Cluj-Napoca, Romania
| | | | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, 1435916471, Iran
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 40015, Cluj-Napoca, Romania; Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. Ion Chiricuta", 400015, Cluj-Napoca, Romania.
| |
Collapse
|
4
|
Erukainure OL, Hafizur RM, Kabir N, Choudhary MI, Atolani O, Banerjee P, Preissner R, Chukwuma CI, Muhammad A, Amonsou EO, Islam MS. Suppressive Effects of Clerodendrum volubile P Beauv. [Labiatae] Methanolic Extract and Its Fractions on Type 2 Diabetes and Its Complications. Front Pharmacol 2018; 9:8. [PMID: 29449808 PMCID: PMC5799276 DOI: 10.3389/fphar.2018.00008] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 01/04/2018] [Indexed: 01/01/2023] Open
Abstract
Type 2 diabetes is the most prominent of all diabetes types, contributing to global morbidity and mortality. Availability and cost of treatment with little or no side effect especially in developing countries, remains a huge burden. This has led to the search of affordable alternative therapies especially from medicinal plants. In this study, the antidiabetic effect of the methanolic extract, dichloromethane (DCM), butanol (BuOH) and aqueous fractions of Clerodendrum volubile leaves were investigated in type 2 diabetic rats for their effect on glucose homeostasis, serum insulin level and hepatic biomarkers, lipid profile, pancreatic redox balance and Ca2+ levels, and β-cell distribution and function. The DCM was further fractionated to isolate the active compounds, biochanin and 5,7,4'-trimethoxykaempferol. They were investigated for their toxicity and ADMET properties, α-glucosidase and angiotensin I converting enzyme (ACE) inhibitory activities in silico. There were significant (p < 0.05) decrease in blood glucose, cholesterol, LDL-C, vLDL-C, triglyceride, AST and ALT levels in all treated groups, with DCM fraction showing the best activity. All treated rats showed significantly (p < 0.05) improved anti-oxidative activities. Treatment with the DCM fraction led to significant (p < 0.05) increased serum insulin and pancreatic Ca2+ levels, as well as improved β-cell distribution and function. DCM fraction also showed improved glucose tolerance. DCM fraction dose-dependently inhibited ACE activity. The toxicity class of the isolated compounds was predicted to be 5. They were also predicted to be potent inhibitors of cytochrome P (CYPs) 1A2, 2D6 and 3A4. They docked well with α-glucosidase and ACE. These results indicate the therapeutic potential of the plant against type 2 diabetes, with the DCM fraction being the most potent which may be attributed to the isolated flavones. It further suggests antihypertensive potentials of the DCM fraction. However, inhibition of CYPs by the flavones may suggest caution in usage with other prescribed drugs metabolized by these enzymes.
Collapse
Affiliation(s)
- Ochuko L. Erukainure
- Nutrition and Toxicology Division, Federal Institute of Industrial Research Oshodi, Lagos, Nigeria
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| | - Rahman M. Hafizur
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Nurul Kabir
- Faculty of Science, Institute of Biological Sciences, University of Malaya, Kuala Lumpur, Malaysia
| | - M. Iqbal Choudhary
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Olubunmi Atolani
- Structural Bioinformatics Group, Institute for Physiology, Charité – University Medicine Berlin, Berlin, Germany
- Department of Chemistry, University of Ilorin, Ilorin, Nigeria
| | - Priyanka Banerjee
- Structural Bioinformatics Group, Institute for Physiology, Charité – University Medicine Berlin, Berlin, Germany
| | - Robert Preissner
- Structural Bioinformatics Group, Institute for Physiology, Charité – University Medicine Berlin, Berlin, Germany
| | - Chika I. Chukwuma
- Department of Food Technology, Durban University of Technology, Steve Biko Campus, Durban, South Africa
- Department of Pharmacology, University of the Free State, Bloemfontein, South Africa
| | - Aliyu Muhammad
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
- Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria
| | - Eric O. Amonsou
- Department of Food Technology, Durban University of Technology, Steve Biko Campus, Durban, South Africa
| | - Md. Shahidul Islam
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| |
Collapse
|
5
|
van Ginkel PR, Yan MB, Bhattacharya S, Polans AS, Kenealey JD. Natural products induce a G protein-mediated calcium pathway activating p53 in cancer cells. Toxicol Appl Pharmacol 2015; 288:453-62. [PMID: 26341291 DOI: 10.1016/j.taap.2015.08.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 08/20/2015] [Accepted: 08/22/2015] [Indexed: 10/23/2022]
Abstract
Paclitaxel, etoposide, vincristine and doxorubicin are examples of natural products being used as chemotherapeutics but with adverse side effects that limit their therapeutic window. Natural products derived from plants and having low toxicity, such as quercetin, resveratrol, epigallocatechin gallate and piceatannol, have been shown to inhibit tumor cell growth both in vitro and in pre-clinical models of cancer, but their mechanisms of action have not been fully elucidated, thus restricting their use as prototypes for developing synthetic analogs with improved anti-cancer properties. We and others have demonstrated that one of the earliest and consistent events upon exposure of tumor cells to these less toxic natural products is a rise in cytoplasmic calcium, activating several pro-apoptotic pathways. We describe here a G protein/inositol 1,4,5-trisphosphate pathway (InsP3) in MDA-MB-231 human breast cancer cells that mediates between these less toxic natural products and the release of calcium from the endoplasmic reticulum. Further, we demonstrate that this elevation of intracellular calcium modulates p53 activity and the subsequent transcription of several pro-apoptotic genes encoding PIG8, CD95, PIDD, TP53INP, RRM2B, Noxa, p21 and PUMA. We conclude from our findings that less toxic natural products likely bind to a G protein coupled receptor that activates a G protein-mediated and calcium-dependent pathway resulting selectively in tumor cell death.
Collapse
Affiliation(s)
- Paul R van Ginkel
- UW Carbone Cancer Center, University of Wisconsin, Madison, WI 53792, United States; Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI 53792, United States
| | - Michael B Yan
- UW Carbone Cancer Center, University of Wisconsin, Madison, WI 53792, United States; Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI 53792, United States
| | - Saswati Bhattacharya
- UW Carbone Cancer Center, University of Wisconsin, Madison, WI 53792, United States; Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI 53792, United States; Department of Pediatrics, University of Wisconsin, Madison, WI 53792, United States
| | - Arthur S Polans
- UW Carbone Cancer Center, University of Wisconsin, Madison, WI 53792, United States; Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI 53792, United States.
| | - Jason D Kenealey
- UW Carbone Cancer Center, University of Wisconsin, Madison, WI 53792, United States; Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI 53792, United States; Department of Nutrition, Dietetics and Food Science, Brigham Young University, Provo, UT 84602, United States
| |
Collapse
|
6
|
Ranzato E, Magnelli V, Martinotti S, Waheed Z, Cain SM, Snutch TP, Marchetti C, Burlando B. Epigallocatechin-3-gallate elicits Ca2+ spike in MCF-7 breast cancer cells: essential role of Cav3.2 channels. Cell Calcium 2014; 56:285-95. [PMID: 25260713 DOI: 10.1016/j.ceca.2014.09.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Revised: 05/29/2014] [Accepted: 09/01/2014] [Indexed: 12/26/2022]
Abstract
We used MCF-7 human breast cancer cells that endogenously express Cav3.1 and Cav3.2 T-type Ca(2+) channels toward a mechanistic study on the effect of EGCG on [Ca(2+)]i. Confocal Ca(2+) imaging showed that EGCG induces a [Ca(2+)]i spike which is due to extracellular Ca(2+) entry and is sensitive to catalase and to low-specificity (mibefradil) and high-specificity (Z944) T-type Ca(2+)channel blockers. siRNA knockdown of T-type Ca(2+) channels indicated the involvement of Cav3.2 but not Cav3.1. Application of EGCG to HEK cells expressing either Cav3.2 or Cav3.1 induced enhancement of Cav3.2 and inhibition of Cav3.1 channel activity. Measurements of K(+) currents in MCF-7 cells showed a reversible, catalase-sensitive inhibitory effect of EGCG, while siRNA for the Kv1.1 K(+) channel induced a reduction of the EGCG [Ca(2+)]i spike. siRNA for Cav3.2 reduced EGCG cytotoxicity to MCF-7 cells, as measured by calcein viability assay. Together, data suggest that EGCG promotes the activation of Cav3.2 channels through K(+) current inhibition leading to membrane depolarization, and in addition increases Cav3.2 currents. Cav3.2 channels are in part responsible for EGCG inhibition of MCF-7 viability, suggesting that deregulation of [Ca(2+)]i by EGCG may be relevant in breast cancer treatment.
Collapse
Affiliation(s)
- Elia Ranzato
- Dipartimento di Scienze e Innovazione Tecnologica, DiSIT, Università del Piemonte Orientale, viale T. Michel 11, 15121 Alessandria, Italy
| | - Valeria Magnelli
- Dipartimento di Scienze e Innovazione Tecnologica, DiSIT, Università del Piemonte Orientale, viale T. Michel 11, 15121 Alessandria, Italy
| | - Simona Martinotti
- Dipartimento di Scienze e Innovazione Tecnologica, DiSIT, Università del Piemonte Orientale, viale T. Michel 11, 15121 Alessandria, Italy
| | - Zeina Waheed
- Michael Smith Laboratories, University of British Columbia, Rm 219 - 2185 East Mall, Vancouver, BC, Canada V6T 1Z4
| | - Stuart M Cain
- Michael Smith Laboratories, University of British Columbia, Rm 219 - 2185 East Mall, Vancouver, BC, Canada V6T 1Z4
| | - Terrance P Snutch
- Michael Smith Laboratories, University of British Columbia, Rm 219 - 2185 East Mall, Vancouver, BC, Canada V6T 1Z4
| | - Carla Marchetti
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, via De Marini 6, 16149 Genova, Italy
| | - Bruno Burlando
- Dipartimento di Scienze e Innovazione Tecnologica, DiSIT, Università del Piemonte Orientale, viale T. Michel 11, 15121 Alessandria, Italy; Istituto di Biofisica, Consiglio Nazionale delle Ricerche, via De Marini 6, 16149 Genova, Italy.
| |
Collapse
|
7
|
Yoon JS, Kim HM, Yadunandam AK, Kim NH, Jung HA, Choi JS, Kim CY, Kim GD. Neferine isolated from Nelumbo nucifera enhances anti-cancer activities in Hep3B cells: molecular mechanisms of cell cycle arrest, ER stress induced apoptosis and anti-angiogenic response. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2013; 20:1013-1022. [PMID: 23746959 DOI: 10.1016/j.phymed.2013.03.024] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 02/19/2013] [Accepted: 03/26/2013] [Indexed: 06/02/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most aggressive malignant diseases and is highly resistant to conventional chemotherapy. Neferine, a major bisbenzylisoquinoline alkaloid derived from the embryos of Nelumbo nucifera, has been reported a few physiological activities. However, the mechanisms of anticancer effects are not well understood and its detailed activities on Hep3B cells have not been determined. Our results suggest that neferine exhibited cytotoxicity against HCC Hep3B cells, but not against HCC Sk-Hep1 and THLE-3, a normal human liver cell line. In addition, consistent with the induction of G1/S phase cell population in flow cytometry, downregulation of c-Myc, cyclin D1, D3, CDK4, E2F-1, as well as dephosphorlyation of cdc2 by western blot analysis, as evidenced by the appearance of cell cycle arrest, were observed in Hep3B cells treated with neferine. Our results demonstrated neferine induced ER stress and apoptosis, acting through multiple signaling cascades by the activation of Bim, Bid, Bax, Bak, Puma, caspases-3, -6, -7, -8 and PARP, and the protein expression levels of Bip, calnexin, PDI, calpain-2 and caspase-12 were also upregulated dramatically by neferine treatment. Overexpression of GFP-LC3B by neferine resulted in a diffuse cytosolic GFP fluorescence and the strong fluorescent spots, representing autophagosomes. The significant reduction of the migration in Hep3B cells and the capillary tube-like formation of HUVECs by neferine were also determined. These observations reveal that the therapeutic potential of neferine in treating HCC Hep3B cells, containing copies of hepatitis B virus (HBV) genomes.
Collapse
Affiliation(s)
- Jin-Soo Yoon
- Department of Microbiology, College of Natural Sciences, Pukyong National University, Namgu, Busan 608-737, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Lamoral-Theys D, Wauthoz N, Heffeter P, Mathieu V, Jungwirth U, Lefranc F, Nève J, Dubois J, Dufrasne F, Amighi K, Berger W, Gailly P, Kiss R. Trivanillic polyphenols with anticancer cytostatic effects through the targeting of multiple kinases and intracellular Ca2+ release. J Cell Mol Med 2012; 16:1421-34. [PMID: 21810170 PMCID: PMC3823212 DOI: 10.1111/j.1582-4934.2011.01403.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Cancer cells exhibit de-regulation of multiple cellular signalling pathways and treatments of various types of cancers with polyphenols are promising. We recently reported the synthesis of a series of 33 novel divanillic and trivanillic polyphenols that displayed anticancer activity, at least in vitro, through inhibiting various kinases. This study revealed that minor chemical modifications of a trivanillate scaffold could convert cytotoxic compounds into cytostatic ones. Compound 13c, a tri-chloro derivative of trivanillic ester, displayed marked inhibitory activities against FGF-, VEGF-, EGF- and Src-related kinases, all of which are implicated not only in angiogenesis but also in the biological aggressiveness of various cancer types. The pan-anti-kinase activity of 13c occurs at less than one-tenth of its mean IC50in vitro growth inhibitory concentrations towards a panel of 12 cancer cell lines. Of the 26 kinases for which 13c inhibited their activity by >75%, eight (Yes, Fyn, FGF-R1, EGFR, Btk, Mink, Ret and Itk) are implicated in control of the actin cytoskeleton organization to varying degrees. Compound 13c accordingly impaired the typical organization of the actin cytoskeleton in human U373 glioblastoma cells. The pan-anti-kinase activity and actin cytoskeleton organization impairment provoked by 13c concomitantly occurs with calcium homeostasis impairment but without provoking MDR phenotype activation. All of these anticancer properties enabled 13c to confer therapeutic benefits in vivo in a mouse melanoma pseudometastatic lung model. These data argue in favour of further chemically modifying trivanillates to produce novel and potent anticancer drugs.
Collapse
Affiliation(s)
- Delphine Lamoral-Theys
- Laboratoire de Chimie BioAnalytique, Toxicologie et Chimie Physique Appliquée, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Ma X, He D, Ru X, Chen Y, Cai Y, Bruce IC, Xia Q, Yao X, Jin J. Apigenin, a plant-derived flavone, activates transient receptor potential vanilloid 4 cation channel. Br J Pharmacol 2012; 166:349-58. [PMID: 22049911 DOI: 10.1111/j.1476-5381.2011.01767.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Transient receptor potential vanilloid 4 (TRPV4) is a Ca(2+) -permeable channel with multiple modes of activation. Apigenin is a plant-derived flavone, which has potential preventive effects on the development of cardiovascular disease. We set out to explore the effects of apigenin on TRPV4 channel activity and its role in vasodilatation. EXPERIMENTAL APPROACH The effects of apigenin (0.01-30 µM) on TPRV4 channels were investigated in HEK293 cells over-expressing TRPV4, rat primary cultured mesenteric artery endothelial cells (MAECs) and isolated small mesenteric arterial segments using whole-cell patch clamp, fluorescent Ca(2+) imaging, intracellular recording and pressure myography. KEY RESULTS Whole-cell patch clamp and fluorescent Ca(2+) imaging in HEK cells over-expressing TRPV4 showed that apigenin concentration-dependently stimulated the TRPV4-mediated cation current and Ca(2+) influx. In MAECs, apigenin stimulated Ca(2+) influx in a concentration-dependent manner. These increases in cation current and Ca(2+) influx were markedly inhibited by TRPV4-specific blockers and siRNAs. Furthermore, pressure myography and intracellular recording in small third-order mesenteric arteries showed that apigenin dose-dependently evoked smooth muscle cell membrane hyperpolarization and subsequent vascular dilatation, which were significantly inhibited by TRPV4-specific blockers. TRPV4 blocker or charybdotoxin (200 nM) plus apamin (100 nM) diminished the apigenin-induced dilatation. CONCLUSION AND IMPLICATIONS This is the first study to demonstrate the selective stimulation of TRPV4 by apigenin. Apigenin was found to activate TRPV4 channels in a dose-dependent manner in HEK cells over-expressing TRPV4 and in native endothelial cells. In rat small mesenteric arteries, apigenin acts on TRPV4 in endothelial cells to induce EDHF-mediated vascular dilatation.
Collapse
Affiliation(s)
- Xin Ma
- Department of Cellular and Molecular Pharmacology, Jiangnan University, Wuxi, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Soler F, Asensio MC, Fernández-Belda F. Inhibition of the intracellular Ca(2+) transporter SERCA (Sarco-Endoplasmic Reticulum Ca(2+)-ATPase) by the natural polyphenol epigallocatechin-3-gallate. J Bioenerg Biomembr 2012; 44:597-605. [PMID: 22851007 DOI: 10.1007/s10863-012-9462-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 07/08/2012] [Indexed: 11/26/2022]
Abstract
The use of a microsomal preparation from skeletal muscle revealed that both Ca(2+) transport and Ca(2+)-dependent ATP hydrolysis linked to Sarco-Endoplasmic Reticulum Ca(2+)-ATPase are inhibited by epigallocatechin-3-gallate (EGCG). A half-maximal effect was achieved at approx. 12 μM. The presence of the galloyl group was essential for the inhibitory effect of the catechin. The relative inhibition of the Ca(2+)-ATPase activity decreased when the Ca(2+) concentration was raised but not when the ATP concentration was elevated. Data on the catalytic cycle indicated inhibition of maximal Ca(2+) binding and a decrease in Ca(2+) binding affinity when measured in the absence of ATP. Moreover, the addition of ATP to samples in the presence of EGCG and Ca(2+) led to an early increase in phosphoenzyme followed by a time-dependent decay that was faster when the drug concentration was raised. However, phosphorylation following the addition of ATP plus Ca(2+) led to a slow rate of phosphoenzyme accumulation that was also dependent on EGCG concentration. The results are consistent with retention of the transporter conformation in the Ca(2+)-free state, thus impeding Ca(2+) binding and therefore the subsequent steps when ATP is added to trigger the Ca(2+) transport process. Furthermore, phosphorylation by inorganic phosphate in the absence of Ca(2+) was partially inhibited by EGCG, suggesting alteration of the native Ca(2+)-free conformation at the catalytic site.
Collapse
Affiliation(s)
- Fernando Soler
- Departamento de Bioquímica y Biología Molecular A, Universidad de Murcia, Campus de Espinardo, Murcia, Spain
| | | | | |
Collapse
|
11
|
Zhang R, Piao MJ, Kim KC, Kim AD, Choi JY, Choi J, Hyun JW. Endoplasmic reticulum stress signaling is involved in silver nanoparticles-induced apoptosis. Int J Biochem Cell Biol 2011; 44:224-32. [PMID: 22064246 DOI: 10.1016/j.biocel.2011.10.019] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 10/19/2011] [Accepted: 10/25/2011] [Indexed: 10/15/2022]
Abstract
Although silver nanoparticles (AgNPs) have been reported to exert strong acute toxic effects on various cultured cells by inducing oxidative stress, the molecular mechanisms by which AgNPs-damaged cells are unknown. Because the endoplasmic reticulum (ER) may play an important role in the response to oxidative stress-induced damage and is quite sensitive to oxidative damage, we hypothesized that AgNPs may exert cytotoxic effects on cells by modulating ER stress. In our study, AgNPs resulted in cytotoxicity and apoptotic cell death when analyzing cell viability, DNA fragmentation and the apoptotic sub-G(1) population. Flow cytometry and confocal microscopy indicated that the cells were sensitive to AgNPs with respect to the induction of mitochondrial Ca(2+) overloading and enhancement of ER stress. AgNPs induced a number of signature ER stress markers, including phosphorylation of RNA-dependent protein kinase-like ER kinase (PERK) and its downstream eukaryotic initiation factor 2α, phosphorylation of inositol-requiring protein 1 (IRE1), splicing of ER stress-specific X-box transcription factor-1, cleavage of activating transcription factor 6 (ATF6) and up-regulation of glucose-regulated protein-78 and CCAAT/enhancer-binding protein-homologous protein (CHOP/GADD153). Down-regulation of PERK, IRE1 and ATF6 expression using siRNA significantly decreased AgNPs-induced the enhancement of ER stress. In addition, down-regulation of CHOP expression with siRNA CHOP attenuated AgNPs-induced apoptosis. Taken together, the present study supports an important role for the ER stress response in mediating AgNPs-induced apoptosis.
Collapse
Affiliation(s)
- Rui Zhang
- School of Medicine and Applied Radiological Science Research Institute, Jeju National University, Jeju, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
12
|
Révész K, Tüttő A, Szelényi P, Konta L. Tea flavan-3-ols as modulating factors in endoplasmic reticulum function. Nutr Res 2011; 31:731-40. [DOI: 10.1016/j.nutres.2011.09.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 09/07/2011] [Accepted: 09/15/2011] [Indexed: 01/04/2023]
|
13
|
Kargacin ME, Emmett TL, Kargacin GJ. Epigallocatechin-3-gallate has dual, independent effects on the cardiac sarcoplasmic reticulum/endoplasmic reticulum Ca2+ ATPase. J Muscle Res Cell Motil 2011; 32:89-98. [PMID: 21818690 DOI: 10.1007/s10974-011-9256-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 07/23/2011] [Indexed: 11/25/2022]
Abstract
We determined the effects of epigallocatechin-3-gallate (EGCG) and epicatechin (EC), on pump turnover and Ca2+ transport by the cardiac form of the sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA). Fluorescence spectroscopy was used to directly measure SERCA ATPase activity and to measure Ca2+ uptake into cardiac sarcoplasmic reticulum (SR) vesicles and microsomes derived from human embryonic kidney (HEK) cells expressing human cardiac SERCA2a. We found that EGCG reduces the maximum velocity of Ca2+ uptake into cardiac SR vesicles and increases the Ca2+-sensitivity of uptake in a concentration-dependent manner. EC is less potent than EGCG in increasing the Ca2+-sensitivity of uptake and does not affect maximum uptake velocity. The EGCG-dependent reduction in Ca2+ uptake velocity is well correlated with direct inhibition of SERCA. The effect of EGCG on the Ca2+-sensitivity of Ca2+ uptake into cardiac SR vesicles is affected by the phosphorylation status of phospholamban (PLB). When cardiac SERCA2a is expressed in HEK cells without PLB, EGCG reduces the maximum velocity of Ca2+ uptake but does not affect the Ca2+-sensitivity of uptake into microsomes derived from these cells indicating that the effect of EGCG on Ca2+-sensitivity requires the presence of PLB. Our results show that EGCG has dual effects on SERCA function in cardiac SR vesicles: it directly affects SERCA by reducing maximum uptake velocity; it increases the Ca2+-sensitivity of Ca2+ uptake in a manner that appears to depend on the interaction between SERCA and PLB.
Collapse
Affiliation(s)
- M E Kargacin
- Department of Physiology and Pharmacology, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | | | | |
Collapse
|
14
|
Ahn SH, Kim HJ, Jeong I, Hong YJ, Kim MJ, Rhie DJ, Jo YH, Hahn SJ, Yoon SH. Grape seed proanthocyanidin extract inhibits glutamate-induced cell death through inhibition of calcium signals and nitric oxide formation in cultured rat hippocampal neurons. BMC Neurosci 2011; 12:78. [PMID: 21810275 PMCID: PMC3160962 DOI: 10.1186/1471-2202-12-78] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 08/03/2011] [Indexed: 11/30/2022] Open
Abstract
Background Proanthocyanidin is a polyphenolic bioflavonoid with known antioxidant activity. Some flavonoids have a modulatory effect on [Ca2+]i. Although proanthocyanidin extract from blueberries reportedly affects Ca2+ buffering capacity, there are no reports on the effects of proanthocyanidin on glutamate-induced [Ca2+]i or cell death. In the present study, the effects of grape seed proanthocyanidin extract (GSPE) on glutamate-induced excitotoxicity was investigated through calcium signals and nitric oxide (NO) in cultured rat hippocampal neurons. Results Pretreatment with GSPE (0.3-10 μg/ml) for 5 min inhibited the [Ca2+]i increase normally induced by treatment with glutamate (100 μM) for 1 min, in a concentration-dependent manner. Pretreatment with GSPE (6 μg/ml) for 5 min significantly decreased the [Ca2+]i increase normally induced by two ionotropic glutamate receptor agonists, N-methyl-D-aspartate and alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA). GSPE further decreased AMPA-induced response in the presence of 1 μM nimodipine. However, GSPE did not affect the 50 mM K+-induced increase in [Ca2+]i. GSPE significantly decreased the metabotropic glutamate receptor agonist (RS)-3,5-Dihydroxyphenylglycine-induced increase in [Ca2+]i, but it did not affect caffeine-induced response. GSPE (0.3-6 μg/ml) significantly inhibited synaptically induced [Ca2+]i spikes by 0.1 mM [Mg2+]o. In addition, pretreatment with GSPE (6 μg/ml) for 5 min inhibited 0.1 mM [Mg2+]o- and glutamate-induced formation of NO. Treatment with GSPE (6 μg/ml) significantly inhibited 0.1 mM [Mg2+]o- and oxygen glucose deprivation-induced neuronal cell death. Conclusions All these data suggest that GSPE inhibits 0.1 mM [Mg2+]o- and oxygen glucose deprivation-induced neurotoxicity through inhibition of calcium signals and NO formation in cultured rat hippocampal neurons.
Collapse
Affiliation(s)
- Seo-Hee Ahn
- Department of Physiology, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul 137-701, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Singh BN, Shankar S, Srivastava RK. Green tea catechin, epigallocatechin-3-gallate (EGCG): mechanisms, perspectives and clinical applications. Biochem Pharmacol 2011; 82:1807-21. [PMID: 21827739 DOI: 10.1016/j.bcp.2011.07.093] [Citation(s) in RCA: 1055] [Impact Index Per Article: 75.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 07/22/2011] [Accepted: 07/25/2011] [Indexed: 12/22/2022]
Abstract
An expanding body of preclinical evidence suggests EGCG, the major catechin found in green tea (Camellia sinensis), has the potential to impact a variety of human diseases. Apparently, EGCG functions as a powerful antioxidant, preventing oxidative damage in healthy cells, but also as an antiangiogenic and antitumor agent and as a modulator of tumor cell response to chemotherapy. Much of the cancer chemopreventive properties of green tea are mediated by EGCG that induces apoptosis and promotes cell growth arrest by altering the expression of cell cycle regulatory proteins, activating killer caspases, and suppressing oncogenic transcription factors and pluripotency maintain factors. In vitro studies have demonstrated that EGCG blocks carcinogenesis by affecting a wide array of signal transduction pathways including JAK/STAT, MAPK, PI3K/AKT, Wnt and Notch. EGCG stimulates telomere fragmentation through inhibiting telomerase activity. Various clinical studies have revealed that treatment by EGCG inhibits tumor incidence and multiplicity in different organ sites such as liver, stomach, skin, lung, mammary gland and colon. Recent work demonstrated that EGCG reduced DNMTs, proteases, and DHFR activities, which would affect transcription of TSGs and protein synthesis. EGCG has great potential in cancer prevention because of its safety, low cost and bioavailability. In this review, we discuss its cancer preventive properties and its mechanism of action at numerous points regulating cancer cell growth, survival, angiogenesis and metastasis. Therefore, non-toxic natural agent could be useful either alone or in combination with conventional therapeutics for the prevention of tumor progression and/or treatment of human malignancies.
Collapse
Affiliation(s)
- Brahma N Singh
- Department of Pharmacology, Toxicology and Therapeutics, and Medicine, The University of Kansas Cancer Center, The University of Kansas Medical Center, Kansas City, KS 66160, United States
| | | | | |
Collapse
|
16
|
Scholz EP, Zitron E, Katus HA, Karle CA. Cardiovascular ion channels as a molecular target of flavonoids. Cardiovasc Ther 2010; 28:e46-52. [PMID: 20633021 DOI: 10.1111/j.1755-5922.2010.00212.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Flavonoids are a class of naturally occurring polyphenols abundant in edibles and beverages of plant origin. Epidemiological studies consistently associate high flavonoid intake with a reduced risk for the development of cardiovascular diseases. So far these beneficial effects have been mainly attributed to nonspecific antioxidant and antiinflammatory properties. However, there is an increasing body of evidence that flavonoids specifically target molecular structures including cardiovascular ion channels. Playing a pivotal role in the regulation of vascular tone and cardiac electric activity, ion channels represent a major target for the induction of antihypertensive and cardioprotective effects. Thus, pharmacological properties of flavonoids on cardiovascular ion channels, ion currents and tissue preparations are being increasingly addressed in experimental studies. Whereas it has become clear that cardiovascular ion channels represent an important molecular target of flavonoids, the published data have not yet been systematically reviewed.
Collapse
Affiliation(s)
- Eberhard P Scholz
- Department of Internal Medicine III (Cardiology), University Hospital Heidelberg, Heidelberg, Germany.
| | | | | | | |
Collapse
|
17
|
Feng W, Cherednichenko G, Ward CW, Padilla IT, Cabrales E, Lopez JR, Eltit JM, Allen PD, Pessah IN. Green tea catechins are potent sensitizers of ryanodine receptor type 1 (RyR1). Biochem Pharmacol 2010; 80:512-21. [PMID: 20471964 PMCID: PMC2907350 DOI: 10.1016/j.bcp.2010.05.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 05/04/2010] [Accepted: 05/07/2010] [Indexed: 01/26/2023]
Abstract
Catechins, polyphenols extracted from green tea leaves, have a broad range of biological activities although the specific molecular mechanisms responsible are not known. At the high experimental concentrations typically used polyphenols bind to membrane phospholipid and also are easily auto-oxidized to generate superoxide anion and semiquinones, and can adduct to protein thiols. We report that the type 1 ryanodine receptor (RyR1) is a molecular target that responds to nanomolar (-)-epigallocatechin-3-gallate (EGCG) and (-)-epicatechin-3-gallate (ECG). Single channel analyses demonstrate EGCG (5-10nM) increases channel open probability (Po) twofold, by lengthening open dwell time. The degree of channel activation is concentration-dependent and is rapidly and fully reversible. Four related catechins, EGCG, ECG, EGC ((-)-epigallocatechin) and EC ((-)-epicatechin) showed a rank order of activity toward RyR1 (EGCG>ECG>>EGC>>>EC). EGCG and ECG enhance the sensitivity of RyR1 to activation by < or =100microM cytoplasmic Ca(2+) without altering inhibitory potency by >100microM Ca(2+). EGCG as high as 10microM in the extracellular medium potentiated Ca(2+) transient amplitudes evoked by electrical stimuli applied to intact myotubes and adult FDB fibers, without eliciting spontaneous Ca(2+) release or slowing Ca(2+) transient recovery. The results identify RyR1 as a sensitive target for the major tea catechins EGCG and ECG, and this interaction is likely to contribute to their observed biological activities.
Collapse
Affiliation(s)
- Wei Feng
- Department of Molecular Biosciences, University of California, Davis, 95616, United States of America.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Guo Y, Hong YJ, Jang HJ, Kim MJ, Rhie DJ, Jo YH, Hahn SJ, Yoon SH. Octyl Gallate Inhibits ATP-induced Intracellular Calcium Increase in PC12 Cells by Inhibiting Multiple Pathways. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2010; 14:21-8. [PMID: 20221276 DOI: 10.4196/kjpp.2010.14.1.21] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Revised: 01/18/2010] [Accepted: 01/26/2010] [Indexed: 01/18/2023]
Abstract
Phenolic compounds affect intracellular free Ca(2+) concentration ([Ca(2+)](i)) signaling. The study examined whether the simple phenolic compound octyl gallate affects ATP-induced Ca(2+) signaling in PC12 cells using fura-2-based digital Ca(2+) imaging and whole-cell patch clamping. Treatment with ATP (100 microM) for 90 s induced increases in [Ca(2+)](i) in PC12 cells. Pretreatment with octyl gallate (100 nM to 20 microM) for 10 min inhibited the ATP-induced [Ca(2+)](i) response in a concentration-dependent manner (IC(50)=2.84 microM). Treatment with octyl gallate (3 microM) for 10 min significantly inhibited the ATP-induced response following the removal of extracellular Ca(2+) with nominally Ca(2+)-free HEPES HBSS or depletion of intracellular Ca(2+) stores with thapsigargin (1 microM). Treatment for 10 min with the L-type Ca(2+) channel antagonist nimodipine (1 microM) significantly inhibited the ATP-induced [Ca(2+)](i) increase, and treatment with octyl gallate further inhibited the ATP-induced response. Treatment with octyl gallate significantly inhibited the [Ca(2+)](i) increase induced by 50 mM KCl. Pretreatment with protein kinase C inhibitors staurosporin (100 nM) and GF109203X (300 nM), or the tyrosine kinase inhibitor genistein (50 microM) did not significantly affect the inhibitory effects of octyl gallate on the ATP-induced response. Treatment with octyl gallate markedly inhibited the ATP-induced currents. Therefore, we conclude that octyl gallate inhibits ATP-induced [Ca(2+)](i) increase in PC12 cells by inhibiting both non-selective P2X receptor-mediated influx of Ca(2+) from extracellular space and P2Y receptor-induced release of Ca(2+) from intracellular stores in protein kinase-independent manner. In addition, octyl gallate inhibits the ATP-induced Ca(2+) responses by inhibiting the secondary activation of voltage-gated Ca(2+) channels.
Collapse
Affiliation(s)
- Yujie Guo
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Elucidation of susceptible factors to endoplasmic reticulum stress-mediated anticancer activity in human hepatocellular carcinoma. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2008; 377:167-77. [PMID: 18228003 DOI: 10.1007/s00210-007-0249-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2007] [Accepted: 12/12/2007] [Indexed: 10/22/2022]
Abstract
The initiation of endoplasmic reticulum (ER) stress has been suggested to play potential roles in hepatocarcinogenesis. However, many obstacles remain as to whether ER stress plays a role in carcinogenesis or tumoricide. This study sought to identify the signals that can serve as anticancer effectors in cells in response to ER stress. Tunicamycin (an N-glycosylation inhibitor) inhibited cell proliferation with IC(50) values of 0.19 and 0.62 microg/ml in hepatoma (Hep) 3B and HepG2 cells, respectively. It induced G1 arrest of the cell cycle in both cell lines. The anticancer mechanism of tunicamycin was investigated in Hep3B cells. Tunicamycin induced a rapid decline of cyclin D1 and cyclin A expression and an early increase of glucose-related protein (GRP) 78 and growth arrest and DNA damage-inducible transcription factor (GADD) 153 levels. Cyclin A was the most sensitive regulator to tunicamycin-triggered degradation mechanism. The association of p27(Kip1) with cyclin D1/cyclin-dependent kinase (Cdk) 4 was also increased by tunicamycin. The inhibition of GADD153 expression by transfection of GADD153 antisense did not modify tunicamycin-induced G1 arrest and cyclin/Cdk expressions. The knockdown of GRP78 expression by the siRNA transfection technique moderately increased tunicamycin-induced apoptosis but not the antiproliferative effect by sulforhodamine B assay. We suggest that tunicamycin induces G1 arrest through down-regulation of cyclins and Cdks, in which cyclin A is more susceptible to ER stress-triggered degradation mechanism in Hep3B cells. The increased association of p27(Kip1) with cyclin D1/Cdk4 may also contribute to tunicamycin-induced cell-cycle arrest. GADD153 and GRP78 play a minor role in tunicamycin-mediated antiproliferative effect, although GRP78 moderately inhibits apoptosis in Hep3B cells. These data provide evidence that cell-cycle regulators are susceptible factors in hepatocellular carcinoma (HCC) responsive to ER stress.
Collapse
|
20
|
Campos-Toimil M, Orallo F. Effects of (-)-epigallocatechin-3-gallate in Ca2+ -permeable non-selective cation channels and voltage-operated Ca2+ channels in vascular smooth muscle cells. Life Sci 2007; 80:2147-2153. [PMID: 17499810 DOI: 10.1016/j.lfs.2007.04.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2007] [Revised: 04/02/2007] [Accepted: 04/09/2007] [Indexed: 11/24/2022]
Abstract
The effects of (-)-epigallocatechin-3-gallate (EGCG), the most abundant catechin of tea, on Ca(2+)-permeable non-selective cation currents (NSCC) and voltage-operated Ca(2+) channels (VOCC) have been investigated in cultured rat aortic smooth muscle cells using the whole-cell voltage-clamp technique. Under the Cs(+)/tetraethylammonium (TEA)-containing internal solution, and in the presence of nifedipine (1 microM), EGCG (30 microM) activated a long-lasting inward current, with a reversal potential (E(rev)) of approximately 0 mV. This current was not significantly altered by the replacement of [Cl(-)](i) or [Cl(-)](o), implying that the inward current was not a chloride channel, but a NSCC. SKF 96365 (30 microM) and Cd(2+) (500 microM) almost completely abolished the EGCG-induced NSCC. A higher dose of EGCG (100 microM) additionally activated a nifedipine-sensitive inward current in the absence of depolarization protocol. EGCG (100 microM) also potentiated a nifedipine-sensitive voltage-dependent Ba(2+)-current during the first 5 min of incubation. However, after > 10 min of incubation with EGCG, this current was significantly inhibited. Our results suggest that EGCG caused a Ca(2+) influx into smooth muscle cells via VOCC (probably L-type) and other SKF-96365- and Cd(2+)-sensitive Ca(2+)-permeable channels. The action described here may be responsible for the contraction induced by EGCG in rat aortic rings and for the rise of the intracellular concentration of Ca(2+) in rat aortic smooth muscle cells evoked by this catechin. On the other hand, the inhibition of VOCC after > 10 min of incubation may be, in part, responsible for the relaxation of rat aorta induced by EGCG.
Collapse
Affiliation(s)
- Manuel Campos-Toimil
- Departamento de Farmacoloxía, Facultade de Farmacia, Universidade de Santiago de Compostela, Campus Universitario Sur, E-15782 Santiago de Compostela (A Coruña), Spain
| | - Francisco Orallo
- Departamento de Farmacoloxía, Facultade de Farmacia, Universidade de Santiago de Compostela, Campus Universitario Sur, E-15782 Santiago de Compostela (A Coruña), Spain.
| |
Collapse
|
21
|
Srinivasan P, Sabitha KE, Shyamaladevi CS. Modulatory efficacy of green tea polyphenols on glycoconjugates and immunological markers in 4-Nitroquinoline 1-oxide-induced oral carcinogenesis-A therapeutic approach. Chem Biol Interact 2006; 162:149-56. [PMID: 16859662 DOI: 10.1016/j.cbi.2006.05.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2005] [Revised: 05/25/2006] [Accepted: 05/30/2006] [Indexed: 10/24/2022]
Abstract
Green tea polyphenols (GTP) has been used as a chemopreventive agent world wide against chemically induced cancer. The present study is aimed to understand the therapeutic action of GTP on glycoconjugates and immunological markers in 4-Nitroquinoline 1-oxide (4-NQO)-induced oral cancer over a period of 30 days at 200mg/kg, p.o., Oral cancer was induced by painting 4-NQO for 8 weeks followed by administration of GTP after 22 weeks, for 30 days. Glycoconjugates such as hexose, hexosamine, sialicacid, fucose and mucoprotein were analysed. Expression of glycoconjugates was examined through histology and SDS-PAGE. Immunological markers such as circulating immune complex and mast cell density were studied. Oral cancer-induced animals showed a significant increase in levels of glycoconjugates and its expression, similar to that observed for immunological markers. Treatment with GTP altered the expression of glycoconjugates as well as immunological markers. The results suggest that GTP modulates both the expression of glycoconjugates and immunological markers resulting in regression of oral cancer.
Collapse
Affiliation(s)
- Periasamy Srinivasan
- Department of Biochemistry, University of Madras, Guindy Campus, Chennai 600025, Tamilnadu, India
| | | | | |
Collapse
|
22
|
Lin YT, Kwon YI, Labbe RG, Shetty K. Inhibition of Helicobacter pylori and associated urease by oregano and cranberry phytochemical synergies. Appl Environ Microbiol 2006; 71:8558-64. [PMID: 16332847 PMCID: PMC1317381 DOI: 10.1128/aem.71.12.8558-8564.2005] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Ulcer-associated dyspepsia is caused by infection with Helicobacter pylori. H. pylori is linked to a majority of peptic ulcers. Antibiotic treatment does not always inhibit or kill H. pylori with potential for antibiotic resistance. The objective of this study was to determine the potential for using phenolic phytochemical extracts to inhibit H. pylori in a laboratory medium. Our approach involved the development of a specific phenolic profile with optimization of different ratios of extract mixtures from oregano and cranberry. Subsequently, antimicrobial activity and antimicrobial-linked urease inhibition ability were evaluated. The results indicated that the antimicrobial activity was greater in extract mixtures than in individual extracts of each species. The results also indicate that the synergistic contribution of oregano and cranberry phenolics may be more important for inhibition than any species-specific phenolic concentration. Further, based on plate assay, the likely mode of action may be through urease inhibition and disruption of energy production by inhibition of proline dehydrogenase at the plasma membrane.
Collapse
Affiliation(s)
- Y T Lin
- Department of Food Science, Chenoweth Laboratory, University of Massachusetts, Amherst, MA 01003, USA
| | | | | | | |
Collapse
|
23
|
Michel MC, König G, Mohr K, Simmet T. Editorial guidelines for manuscripts on the pharmacology of plant extracts. Naunyn Schmiedebergs Arch Pharmacol 2005; 371:349-50. [PMID: 16007459 DOI: 10.1007/s00210-005-1071-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|