1
|
Chitcholtan K, Singh M, Tino A, Garrill A, Sykes P. Effects of Resveratrol on In Vivo Ovarian Cancer Cells Implanted on the Chorioallantoic Membrane (CAM) of a Chicken Embryo Model. Int J Mol Sci 2024; 25:4374. [PMID: 38673959 PMCID: PMC11049836 DOI: 10.3390/ijms25084374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Ovarian cancer poses a significant threat to patients in its advanced stages, often with limited treatment options available. In such cases, palliative management becomes the primary approach to maintaining a reasonable quality of life. Therefore, the administration of any medication that can benefit patients without a curative option holds potential. Resveratrol, a natural compound known for its in vitro anticancer activities, has generated contrasting results in vivo and human studies. In this study, we aimed to assess the anticancer effects of resveratrol on ovarian cancer cells grown on the chorioallantoic membrane (CAM) of chicken embryos. Two ovarian cancer cell lines, OVCAR-8 and SKOV-3, were cultured in collagen scaffolds for four days before being implanted on the CAM of chicken embryos on day 7. Different doses of resveratrol were applied to the CAM every two days for six days. Subsequently, CAM tissues were excised, fixed, and subjected to histological analysis. Some CAM tumours were extracted to analyse proteins through Western blotting. Our findings indicate that specific doses of resveratrol significantly reduce angiogenic activities, pNF-κB levels, and SLUG protein levels by using immunohistochemistry. These results suggest that resveratrol may have the potential to impact the behaviour of ovarian cancer CAM tumours, thereby warranting further consideration as a complementary treatment option for women with incurable ovarian cancer.
Collapse
Affiliation(s)
- Kenny Chitcholtan
- Gynaecological Cancer Research Group, Department of Obstetrics and Gynaecology, University of Otago Christchurch, Christchurch 8011, New Zealand; (A.T.); (P.S.)
| | - Melanie Singh
- School of Biological Sciences, University of Canterbury, Christchurch 8140, New Zealand; (M.S.); (A.G.)
| | - Alex Tino
- Gynaecological Cancer Research Group, Department of Obstetrics and Gynaecology, University of Otago Christchurch, Christchurch 8011, New Zealand; (A.T.); (P.S.)
| | - Ashley Garrill
- School of Biological Sciences, University of Canterbury, Christchurch 8140, New Zealand; (M.S.); (A.G.)
| | - Peter Sykes
- Gynaecological Cancer Research Group, Department of Obstetrics and Gynaecology, University of Otago Christchurch, Christchurch 8011, New Zealand; (A.T.); (P.S.)
| |
Collapse
|
2
|
Insight into autophagy in platinum resistance of cancer. Int J Clin Oncol 2023; 28:354-362. [PMID: 36705869 DOI: 10.1007/s10147-023-02301-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 01/16/2023] [Indexed: 01/28/2023]
Abstract
Platinum drugs, as a class of widely used chemotherapy agents, frequently appear in the treatment of cancer at different phrases. However, platinum resistance is the major bottleneck of platinum drugs for exerting anti-tumor effect. At present, the mechanism of platinum resistance has been thoroughly explored in terms of drug delivery methods, DNA damage repair function, etc., but it has not yet been translated into an effective weapon for reversing platinum resistance. Recently, autophagy has been proved to be closely related to platinum resistance, and the involved molecular mechanism may provide a new perspective on platinum resistance. The aim of this review is to sort out the studies related to autophagy and platinum resistance, and to focus on summarizing the relevant molecular mechanisms, so as to provide clues for future studies related to autophagy and platinum resistance.
Collapse
|
3
|
Zhang W, Zhang R, Chang Z, Wang X. Resveratrol activates CD8+ T cells through IL-18 bystander activation in lung adenocarcinoma. Front Pharmacol 2022; 13:1031438. [PMCID: PMC9630476 DOI: 10.3389/fphar.2022.1031438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
Resveratrol, a natural product, has demonstrated anti-tumor effects in various kinds of tumor types, including colon, breast, and pancreatic cancers. Most research has focused on the inhibitory effects of resveratrol on tumor cells themselves rather than resveratrol’s effects on tumor immunology. In this study, we found that resveratrol inhibited the growth of lung adenocarcinoma in a subcutaneous tumor model by using the β-cyclodextrin-resveratrol inclusion complex. After resveratrol treatment, the proportion of M2-like tumor-associated macrophages (TAMs) was reduced and tumor-infiltrating CD8T cells showed significantly increased activation. The results of co-culture and antibody neutralization experiments suggested that macrophage-derived IL-18 may be a key cytokine in the resveratrol anti-tumor effect of CD8T cell activation. The results of this study demonstrate a novel view of the mechanisms of resveratrol tumor suppression. This natural product could reprogram TAMs and CD8T effector cells for tumor treatment.
Collapse
Affiliation(s)
- Wei Zhang
- Emergency and Disaster Medical Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Ruohao Zhang
- School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Zhiguang Chang
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- *Correspondence: Zhiguang Chang, ; Xiaobo Wang,
| | - Xiaobo Wang
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- *Correspondence: Zhiguang Chang, ; Xiaobo Wang,
| |
Collapse
|
4
|
The role of resveratrol, Sirtuin1 and RXRα as prognostic markers in ovarian cancer. Arch Gynecol Obstet 2021; 305:1559-1572. [PMID: 34870752 PMCID: PMC9166836 DOI: 10.1007/s00404-021-06262-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 09/15/2021] [Indexed: 11/11/2022]
Abstract
Objective Ovarian cancer is the most lethal gynecologic cancer. Resveratrol (RSV) is known to alter metabolism in cancer. It affects the nuclear retinoid-X-receptor (RXR), which implies a modulating effect of RXR to gynaecologic cancers. Furthermore, RSV targets Sirtuin1 (Sirt1), a histone deacetylase. Study design 123 tissue samples of patients with serous or mucinous ovarian cancer were examined for expression of Sirt1 and RXR. Ovarian cell lines were treated with RSV and consequences on viability and apoptosis were evaluated. The influence of RSV to Sirt1 and RXR expression was analyzed by western blotting Results A correlation of nuclear Sirt1 and RXRα expression could be detected (p = 0.006). Co-expression of nuclear RXRα and cytoplasmic (p = 0.026) or nuclear (p = 0.041) Sirt1 was associated with significantly increased overall survival in advanced tumour stages. Viability was decreased in all cell lines after stimulation with resveratrol, while cell apoptosis was increased. RSV treatment led to significant lower Sirt1 expression in A2780 cells (p = 0.025) and significant increased RXR expression in cisA2780 cells (p = 0.012) Conclusion In order to use RSV as medical target, studies could be developed to improve the understanding of drug resistance mechanisms and consequently improve treatment outcome. Supplementary Information The online version contains supplementary material available at 10.1007/s00404-021-06262-w.
Collapse
|
5
|
Fukui M, Choi HJ, Wang P, Zhu BT. Mechanism underlying resveratrol's attenuation of paclitaxel cytotoxicity in human breast cancer cells: Role of the SIRT1-FOXO1-HER3 signaling pathway. Cancer Treat Res Commun 2021; 28:100386. [PMID: 34010730 DOI: 10.1016/j.ctarc.2021.100386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 04/14/2021] [Accepted: 05/02/2021] [Indexed: 01/14/2023]
Abstract
Resveratrol (RES), a dietary phenolic compound, was reported to have cancer chemoprotective and chemotherapeutic effects. Earlier we unexpectedly observed that RES has a growth-enhancing effect in some breast cancer cells and can diminish the susceptibility of MDA-MB-231 and SKBR-3 cells to paclitaxel-induced cell death, but this phenomenon is not observed in MCF-7 cells. The present study seeks to determine the mechanism underlying RES's attenuation of paclitaxel cytotoxicity in cancer cells. It is found that RES reduces the anticancer action of paclitaxel only in the human breast cancer cells that express HER3 protein. Treatment of SKBR-3 cells with RES increases HER3 expression in a dose-dependent manner. The induction of HER3 expression by RES confers resistance of breast cancer cells against paclitaxel cytotoxicity. Furthermore, it is observed that the SIRT1-FOXO1 signaling pathway plays an important role in mediating RES-induced upregulation of HER3 expression. In conclusion, the present study reveals the mechanism for RES-induced resistance against paclitaxel in some human breast cancer cells, and it is suggested that the combined use of RES and paclitaxel is not suitable for treating human breast cancer that expresses HER3 protein.
Collapse
Affiliation(s)
- Masayuki Fukui
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Hye Joung Choi
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Pan Wang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China; Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China; Shenzhen Bay Laboratory, Shenzhen 518055, China.
| |
Collapse
|
6
|
Chen J, Chen H, Pan L. SIRT1 and gynecological malignancies (Review). Oncol Rep 2021; 45:43. [PMID: 33649834 PMCID: PMC7934219 DOI: 10.3892/or.2021.7994] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 01/25/2021] [Indexed: 12/15/2022] Open
Abstract
Sirtuin 1 (SIRT1), a member of the sirtuin protein family, is a nicotinamide adenine dinucleotide (NAD+)-dependent type III histone deacetylase and mono-ADP-ribosyltransferase. SIRT1 can deacetylate histones (H1, H3, and H4) and non-histone proteins, and it is widely involved in various physiological and pathological processes in the body, including metabolism, aging, transcription, DNA damage and repair, apoptosis, cell cycle regulation, inflammation and cancer. Research has shown that SIRT1 is involved in tumorigenesis, tumor metastasis and chemotherapy resistance, but it exerts opposing effects and plays different roles in different pathogenic processes. Recent studies have demonstrated that SIRT1 may be implicated in the pathogenesis, development, treatment and prognosis of tumors; however, its role in gynecological tumors remains elusive. The aim of the present review was to summarize the pathogenic roles of SIRT1 in cancer, and to provide what is, to the best of our knowledge, the first review of recent advances involving SIRT1 in cervical cancer, endometrial cancer (EC) and ovarian cancer (OC). In addition, the critical research gaps regarding SIRT1, particularly its potential involvement in the concurrence of EC and cervical cancer and its antagonistic effect against poly(ADP-ribose) polymerase inhibitors in OC, were highlighted.
Collapse
Affiliation(s)
- Jiayu Chen
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Houzao Chen
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Lingya Pan
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| |
Collapse
|
7
|
Cocetta V, Quagliariello V, Fiorica F, Berretta M, Montopoli M. Resveratrol as Chemosensitizer Agent: State of Art and Future Perspectives. Int J Mol Sci 2021; 22:2049. [PMID: 33669559 PMCID: PMC7922064 DOI: 10.3390/ijms22042049] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/13/2021] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
Resistance to chemotherapy still remains a major challenge in the clinic, impairing the quality of life and survival rate of patients. The identification of unconventional chemosensitizing agents is therefore an interesting aspect of cancer research. Resveratrol has emerged in the last decades as a fascinating molecule, able to modulate several cancer-related molecular mechanisms, suggesting a possible application as an adjuvant in cancer management. This review goes deep into the existing literature concerning the possible chemosensitizing effect of resveratrol associated with the most conventional chemotherapeutic drugs. Despite the promising effects observed in different cancer types in in vitro studies, the clinical translation still presents strong limitations due to the low bioavailability of resveratrol. Recently, efforts have been moved in the field of drug delivery to identifying possible strategies/formulations useful for a more effective administration. Despite the necessity of a huge implementation in this research area, resveratrol appears as a promising molecule able to sensitize resistant tumors to drugs, suggesting its potential use in therapy-refractory cancer patients.
Collapse
Affiliation(s)
- Veronica Cocetta
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy;
| | - Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy;
| | - Francesco Fiorica
- Department of Radiation Oncology, Az. ULSS 9 Scaligera, 37045 Legnago, VR, Italy;
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Monica Montopoli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy;
- Veneto Institute of Molecular Medicine, VIMM, 35129 Padova, Italy
| |
Collapse
|
8
|
Ding H, Yu X, Hang C, Gao K, Lao X, Jia Y, Yan Z. Ailanthone: A novel potential drug for treating human cancer. Oncol Lett 2020; 20:1489-1503. [PMID: 32724391 PMCID: PMC7377054 DOI: 10.3892/ol.2020.11710] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 05/05/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer is the second leading cause of death after cardiovascular disease. In 2015, >8.7 million people died worldwide due to cancer, and by 2030 this figure is expected to increase to ~13.1 million. Tumor chemotherapy drugs have specific toxicity and side effects, and patients can also develop secondary drug resistance. To prevent and treat cancer, scientists have developed novel drugs with improved antitumor effects and decreased toxicity. Ailanthone (AIL) is a quassinoid extract from the traditional Chinese medicine plant Ailanthus altissima, which is known to have anti-inflammatory and antimalarial effects. An increasing number of studies have focused on AIL due to its antitumor activity. AIL can inhibit cell proliferation and induce apoptosis by up- or downregulating cancer-associated molecules, which ultimately leads to cancer cell death. Antitumor effects of AIL have been observed in melanoma, acute myeloid leukemia, bladder, lung, breast, gastric and prostate cancer and vestibular neurilemmoma. To the best of our knowledge, the present study is the first review to describe the antitumor mechanisms of AIL.
Collapse
Affiliation(s)
- Haixiang Ding
- Medical School of Ningbo University, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xiuchong Yu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of The Medical School of Ningbo University and Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Chen Hang
- Medical School of Ningbo University, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Kaijun Gao
- Medical School of Ningbo University, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xifeng Lao
- Medical School of Ningbo University, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Yangtao Jia
- Medical School of Ningbo University, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Zhilong Yan
- Department of Gastrointestinal Surgery, The Affiliated Hospital of The Medical School of Ningbo University and Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| |
Collapse
|
9
|
Kim S, Kim W, Kim DH, Jang JH, Kim SJ, Park SA, Hahn H, Han BW, Na HK, Chun KS, Choi BY, Surh YJ. Resveratrol suppresses gastric cancer cell proliferation and survival through inhibition of PIM-1 kinase activity. Arch Biochem Biophys 2020; 689:108413. [PMID: 32473133 DOI: 10.1016/j.abb.2020.108413] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 05/01/2020] [Accepted: 05/17/2020] [Indexed: 02/06/2023]
Abstract
The proviral integration site for Moloney murine leukemia virus (PIM) family of serine/threonine-specific kinases consist of three isoforms, that regulate proliferation, apoptosis, metabolism, invasion, and metastasis of cancer cells. Among these, abnormally elevated kinase activity of PIM-1 contributes to the progression of gastric cancer and predicts poor prognosis and a low survival rate in gastric cancer patients. In the present study, we found that resveratrol, one of the representative chemopreventive and anticarcinogenic phytochemicals, directly binds to PIM-1 and thereby inhibits its catalytic activity in human gastric cancer SNU-601 cells. This resulted in suppression of phosphorylation of the proapoptotic Bad, a known substrate of PIM-1. Resveratrol, by inactivating PIM-1, also inhibited anchorage-independent growth and proliferation of SNU-601 cells. To understand the molecular interaction between resveratrol and PIM-1, we conducted docking simulation and found that resveratrol directly binds to the PIM-1 at the ATP-binding pocket. In conclusion, the proapototic and anti-proliferative effects of resveratrol in gastric cancer cells are likely to be mediated through suppression of PIM-1 kinase activity, which may represent a novel mechanism underlying its chemopreventive and anticarcinogenic actions.
Collapse
Affiliation(s)
- Sujin Kim
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, South Korea; Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Wonki Kim
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Do-Hee Kim
- Department of Chemistry, College of Convergence and Integrated Science, Kyonggi University, Suwon, Gyeonggi-do 16227, South Korea
| | - Jeong-Hoon Jang
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Su-Jung Kim
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Sin-Aye Park
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan 31538, South Korea
| | - Hyunggu Hahn
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Byung Woo Han
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Hye-Kyung Na
- Department of Food Science and Biotechnology, College of Knowledge-based Services Engineering, Sungshin Women's University, Seoul 01133, South Korea
| | - Kyung-Soo Chun
- Department of Pharmacy, College of Pharmacy, Keimyung University, Daegu 42601, South Korea
| | - Bu Young Choi
- Department of Pharmaceutical Science and Engineering, Seowon University, Cheongju, Chungbuk 28674, South Korea.
| | - Young-Joon Surh
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, South Korea; Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Cancer Research Institute, Seoul National University, Seoul 03080, South Korea.
| |
Collapse
|
10
|
Liu Z, Peng Q, Li Y, Gao Y. Resveratrol enhances cisplatin-induced apoptosis in human hepatoma cells via glutamine metabolism inhibition. BMB Rep 2018. [PMID: 30103844 PMCID: PMC6177506 DOI: 10.5483/bmbrep.2018.51.9.114] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cisplatin is one of the most effective chemotherapeutic drugs used in the treatment of HCC, but many patients will ultimately relapse with cisplatin-resistant disease. Used in combination with cisplatin, resveratrol has synergistic effect of increasing chemosensitivity of cisplatin in various cancer cells. However, the mechanisms of resveratrol enhancing cisplatin-induced toxicity have not been well characterized. Our study showed that resveratrol enhances cisplatin toxicity in human hepatoma cells via an apoptosis-dependent mechanism. Further studies reveal that resveratrol decreases the absorption of glutamine and glutathione content by reducing the expression of glutamine transporter ASCT2. Flow cytometric analyses demonstrate that resveratrol and cisplatin combined treatment leads to a significant increase in ROS production compared to resveratrol or cisplatin treated hepatoma cells alone. Phosphorylated H2AX (γH2AX) foci assay demonstrate that both resveratrol and cisplatin treatment result in a significant increase of γH2AX foci in hepatoma cells, and the resveratrol and cisplatin combined treatment results in much more γH2AX foci formation than either resveratrol or cisplatin treatment alone. Furthermore, our studies show that over-expression of ASCT2 can attenuate cisplatin-induced ROS production, γH2AX foci formation and apoptosis in human hepatoma cells. Collectively, our studies suggest resveratrol may sensitize human hepatoma cells to cisplatin chemotherapy via glutamine metabolism inhibition.
Collapse
Affiliation(s)
- Zhaoyuan Liu
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, China
| | - Qing Peng
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, China
| | - Yang Li
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, China
| | - Yi Gao
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province; State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
11
|
Gorga A, Rindone GM, Regueira M, Pellizzari EH, Camberos MC, Cigorraga SB, Riera MF, Galardo MN, Meroni SB. Effect of resveratrol on Sertoli cell proliferation. J Cell Biochem 2018; 119:10131-10142. [DOI: 10.1002/jcb.27350] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 06/28/2018] [Indexed: 01/02/2023]
Affiliation(s)
- A Gorga
- Centro de Investigaciones Endocrinológicas, “Dr César Bergadá,” CONICET‐FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez Buenos Aires Argentina
| | - GM Rindone
- Centro de Investigaciones Endocrinológicas, “Dr César Bergadá,” CONICET‐FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez Buenos Aires Argentina
| | - M Regueira
- Centro de Investigaciones Endocrinológicas, “Dr César Bergadá,” CONICET‐FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez Buenos Aires Argentina
| | - EH Pellizzari
- Centro de Investigaciones Endocrinológicas, “Dr César Bergadá,” CONICET‐FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez Buenos Aires Argentina
| | - MC Camberos
- Centro de Investigaciones Endocrinológicas, “Dr César Bergadá,” CONICET‐FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez Buenos Aires Argentina
| | - SB Cigorraga
- Centro de Investigaciones Endocrinológicas, “Dr César Bergadá,” CONICET‐FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez Buenos Aires Argentina
| | - MF Riera
- Centro de Investigaciones Endocrinológicas, “Dr César Bergadá,” CONICET‐FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez Buenos Aires Argentina
| | - MN Galardo
- Centro de Investigaciones Endocrinológicas, “Dr César Bergadá,” CONICET‐FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez Buenos Aires Argentina
| | - SB Meroni
- Centro de Investigaciones Endocrinológicas, “Dr César Bergadá,” CONICET‐FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez Buenos Aires Argentina
| |
Collapse
|
12
|
Cheng Y, Chen G, Wang L, Kong J, Pan J, Xi Y, Shen F, Huang Z. Triptolide-induced mitochondrial damage dysregulates fatty acid metabolism in mouse sertoli cells. Toxicol Lett 2018; 292:136-150. [PMID: 29723566 DOI: 10.1016/j.toxlet.2018.04.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 04/09/2018] [Accepted: 04/30/2018] [Indexed: 10/17/2022]
Abstract
Triptolide is a major active ingredient of tripterygium glycosides, used for the therapy of immune and inflammatory diseases. However, its clinical applications are limited by severe male fertility toxicity associated with decreased sperm count, mobility and testicular injures. In this study, we determined that triptoide-induced mitochondrial dysfunction triggered reduction of lactate and dysregulation of fatty acid metabolism in mouse Sertoli cells. First, triptolide induced mitochondrial damage through the suppressing of proliferator-activated receptor coactivator-1 alpha (PGC-1α) activity and protein. Second, mitochondrial damage decreased lactate production and dysregulated fatty acid metabolism. Finally, mitochondrial dysfunction was initiated by the inhibition of sirtuin 1 (SIRT1) with the regulation of AMP-activated protein kinase (AMPK) in Sertoli cells after triptolide treatment. Meanwhile, triptolide induced mitochondrial fatty acid oxidation dysregulation by increasing AMPK phosphorylation. Taken together, we provide evidence that the mechanism of triptolide-induced testicular toxicity under mitochondrial injury may involve a metabolic change.
Collapse
Affiliation(s)
- Yisen Cheng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Gaojian Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China.
| | - Li Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China.
| | - Jiamin Kong
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China.
| | - Ji Pan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China.
| | - Yue Xi
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China.
| | - Feihai Shen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China.
| | - Zhiying Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China.
| |
Collapse
|
13
|
Chao SC, Chen YJ, Huang KH, Kuo KL, Yang TH, Huang KY, Wang CC, Tang CH, Yang RS, Liu SH. Induction of sirtuin-1 signaling by resveratrol induces human chondrosarcoma cell apoptosis and exhibits antitumor activity. Sci Rep 2017; 7:3180. [PMID: 28600541 PMCID: PMC5466619 DOI: 10.1038/s41598-017-03635-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 05/02/2017] [Indexed: 02/06/2023] Open
Abstract
Chondrosarcoma is a malignant primary bone tumor. Sirtuin-1 (SIRT1), which is a member of sirtuin family, plays a dual role either in cancer promotion or suppression. There is no report about the role of SIRT1 in the human chondrosarcoma cells. Resveratrol is a potent activator of SIRT1. However, its effects on chondrosarcoma have not been extensively studied. Here, we investigated the role of SIRT1 induction by resveratrol in human chondrosarcoma cell growth and tumor progression. Resveratrol significantly decreased cell viability and induced cell apoptosis in human chondrosarcoma cells in a dose-dependent manner. The protein expression and activity of SIRT1 were activated after treatment with resveratrol. Resveratrol significantly inhibited NF-κB signaling by deacetylating the p65 subunit of NF-κB complex, which could be reversed by siRNA-SIRT1 transfection or deacetylation inhibitor MS-275. Resveratrol induced-apoptosis involved a caspase-3-mediated mechanism. Both siRNA-SIRT1 transfection and MS-275 significantly inhibited the resveratrol-induced caspase-3 cleavage and activity in human chondrosarcoma cells. Moreover, in vivo chondrosarcoma xenograft study revealed a dramatic reduction in tumor volume and the increased SIRT1 and cleaved caspase-3 expressions in tumors by resveratrol treatment. These results suggest that resveratrol induces chondrosarcoma cell apoptosis via a SIRT1-activated NF-κB deacetylation and exhibits anti-chondrosarcoma activity in vivo.
Collapse
Affiliation(s)
- Sung-Chuan Chao
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Surgery, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Ying-Ju Chen
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kuo-How Huang
- Urology, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| | - Kuan-Lin Kuo
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Urology, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| | - Ting-Hua Yang
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
| | - Kuo-Yuan Huang
- Department of Orthopaedics, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Ching-Chia Wang
- Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Rong-Sen Yang
- Department of Orthopaedics, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan. .,Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan. .,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
| |
Collapse
|
14
|
Chen X, Zhang L, Ding S, Lei Q, Fang W. Cisplatin combination drugs induce autophagy in HeLa cells and interact with HSA via electrostatic binding affinity. RSC Adv 2017. [DOI: 10.1039/c7ra00056a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Cisplatin combination drugs induce autophagy in HeLa cells and interact with HSAviaelectrostatic binding affinity.
Collapse
Affiliation(s)
- Xuerui Chen
- Department of Chemistry
- Zhejiang University
- Hangzhou 310027
- China
| | - Li Zhang
- Department of Chemistry
- Zhejiang University
- Hangzhou 310027
- China
| | - Shiping Ding
- School of Medicine
- Zhejiang University
- Hangzhou 310058
- China
| | - Qunfang Lei
- Department of Chemistry
- Zhejiang University
- Hangzhou 310027
- China
| | - Wenjun Fang
- Department of Chemistry
- Zhejiang University
- Hangzhou 310027
- China
| |
Collapse
|
15
|
Pavan AR, Silva GDBD, Jornada DH, Chiba DE, Fernandes GFDS, Man Chin C, Dos Santos JL. Unraveling the Anticancer Effect of Curcumin and Resveratrol. Nutrients 2016; 8:nu8110628. [PMID: 27834913 PMCID: PMC5133053 DOI: 10.3390/nu8110628] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 09/24/2016] [Accepted: 09/27/2016] [Indexed: 12/16/2022] Open
Abstract
Resveratrol and curcumin are natural products with important therapeutic properties useful to treat several human diseases, including cancer. In the last years, the number of studies describing the effect of both polyphenols against cancer has increased; however, the mechanism of action in all of those cases is not completely comprehended. The unspecific effect and the ability to interfere in assays by both polyphenols make this challenge even more difficult. Herein, we analyzed the anticancer activity of resveratrol and curcumin reported in the literature in the last 11 years, in order to unravel the molecular mechanism of action of both compounds. Molecular targets and cellular pathways will be described. Furthermore, we also discussed the ability of these natural products act as chemopreventive and its use in association with other anticancer drugs.
Collapse
Affiliation(s)
- Aline Renata Pavan
- School of Pharmaceutical Sciences, UNESP-Univ Estadual Paulista, Araraquara 14800903, Brazil.
| | | | | | - Diego Eidy Chiba
- School of Pharmaceutical Sciences, UNESP-Univ Estadual Paulista, Araraquara 14800903, Brazil.
| | | | - Chung Man Chin
- School of Pharmaceutical Sciences, UNESP-Univ Estadual Paulista, Araraquara 14800903, Brazil.
| | - Jean Leandro Dos Santos
- School of Pharmaceutical Sciences, UNESP-Univ Estadual Paulista, Araraquara 14800903, Brazil.
| |
Collapse
|
16
|
Lee YJ, Lee GJ, Yi SS, Heo SH, Park CR, Nam HS, Cho MK, Lee SH. Cisplatin and resveratrol induce apoptosis and autophagy following oxidative stress in malignant mesothelioma cells. Food Chem Toxicol 2016; 97:96-107. [PMID: 27591926 DOI: 10.1016/j.fct.2016.08.033] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 08/18/2016] [Accepted: 08/29/2016] [Indexed: 11/25/2022]
Abstract
Malignant mesothelioma (MM) is characterized by poor responsiveness to current chemotherapeutic drugs, usually owing to high resistance to apoptosis. Here, we investigated chemosensitizing effects of phytochemical resveratrol, in combination with cisplatin, on MM cells. The combination treatment of cisplatin and resveratrol (CDDP/RSV) synergistically induced apoptosis, as evidenced by typical cell morphological changes, the appearance of sub-G0/G1 peak, an increase in the Annexin V(+) cells and the cleavage of caspase-3 and PARP. CDDP/RSV increased ROS production and depolarization of mitochondrial membrane potential with an increase in the Bax/Bcl-2 ratio. These changes were attenuated by pretreatment with N-acetylcysteine, suggesting that CDDP/RSV induced apoptosis through oxidative mitochondrial damage. Compared with MSTO-211H cells, CDDP/RSV was less efficient in killing H-2452 cells. H-2452 cells exhibited an enhanced autophagy to CDDP/RSV, as observed by an increase in viable cells exhibiting intense LysoTracker Red staining and up-regulation of Beclin-1 and LC3A. Inhibition of autophagy by bafilomycin A1 rendered cells more sensitive to CDDP/RSV-induced cytotoxicity and this was associated with induction of apoptosis. These data indicate that the increased resistance of H-2452 cells to CDDP/RSV is closely related to the activation of self-defensive autophagy, and provide the rationale for targeting the autophagy regulation in the treatment of MM.
Collapse
Affiliation(s)
- Yoon-Jin Lee
- Department of Biochemistry, College of Medicine, Soonchunhyang University, Cheonan, 330-930, Republic of Korea; Division of Molecular Cancer Research, Soonchunhyang Medical Research Institute, Soonchunhyang University, Cheonan, 330-930, Republic of Korea
| | - Gina J Lee
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, MA, 02115-5000, USA
| | - Sun Shin Yi
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan, 336-745, Republic of Korea
| | - Su-Hak Heo
- R&D Center, C.L. Pharm Co., Ltd., Seongdong-Gu, Seoul 04788, Republic of Korea
| | - Cho-Rong Park
- Department of Biochemistry, College of Medicine, Soonchunhyang University, Cheonan, 330-930, Republic of Korea
| | - Hae-Seon Nam
- Division of Molecular Cancer Research, Soonchunhyang Medical Research Institute, Soonchunhyang University, Cheonan, 330-930, Republic of Korea
| | - Moon-Kyun Cho
- Division of Molecular Cancer Research, Soonchunhyang Medical Research Institute, Soonchunhyang University, Cheonan, 330-930, Republic of Korea
| | - Sang-Han Lee
- Department of Biochemistry, College of Medicine, Soonchunhyang University, Cheonan, 330-930, Republic of Korea.
| |
Collapse
|
17
|
Zulueta A, Caretti A, Signorelli P, Ghidoni R. Resveratrol: A potential challenger against gastric cancer. World J Gastroenterol 2015; 21:10636-10643. [PMID: 26457023 PMCID: PMC4588085 DOI: 10.3748/wjg.v21.i37.10636] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/09/2015] [Accepted: 08/31/2015] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is the fourth most common cancer and the second leading cause of cancer-related mortality in the world. Late diagnosis and classical therapeutic approaches such as surgery, chemotherapy and radiotherapy make this disease a still threatening tumor. Genetic asset, environmental stress, dietary habit and infections caused by Helicobacter pylori (H. pylori) are the major causes concurring to GC initiation. A common mechanism is induction of radicals resulting in gastric mucosal injury. A regular food intake of antioxidant and radical scavenging agents has been proposed to exert protection against tumorigenesis. Resveratrol belongs to the polyphenol flavonoids class of antioxidants produced by a restricted number of plants. Resveratrol exerts bactericidal activity against H. pylori and is a powerful antioxidant, thus acting as a tumor preventive agent. Resveratrol intracellular signaling results in growth arrest and apoptosis, so that it can be directed against tumor progression. Resveratrol therapeutic potential against GC initiation and progression are reviewed here.
Collapse
|
18
|
Tang PCT, Ng YF, Ho S, Gyda M, Chan SW. Resveratrol and cardiovascular health--promising therapeutic or hopeless illusion? Pharmacol Res 2014; 90:88-115. [PMID: 25151891 DOI: 10.1016/j.phrs.2014.08.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 07/29/2014] [Accepted: 08/02/2014] [Indexed: 02/07/2023]
Abstract
Resveratrol (3,5,4'-trihydroxy-trans-stilbene) is a natural polyphenolic compound that exists in Polygonum cuspidatum, grapes, peanuts and berries, as well as their manufactured products, especially red wine. Resveratrol is a pharmacologically active compound that interacts with multiple targets in a variety of cardiovascular disease models to exert protective effects or induce a reduction in cardiovascular risks parameters. This review attempts to primarily serve to summarize the current research findings regarding the putative cardioprotective effects of resveratrol and the molecular pathways underlying these effects. One intent is to hopefully provide a relatively comprehensive resource for clues that may prompt ideas for additional mechanistic studies which might further elucidate and strengthen the role of the stilbene family of compounds in cardiovascular disease and cardioprotection. Model systems that incorporate a significant functional association with tissues outside of the cardiovascular system proper, such as adipose (cell culture, obesity models) and pancreatic (diabetes) tissues, were reviewed, and the molecular pathways and/or targets related to these models and influenced by resveratrol are discussed. Because the body of work encompassing the stilbenes and other phytochemicals in the context of longevity and the ability to presumably mitigate a plethora of afflictions is replete with conflicting information and controversy, especially so with respect to the human response, we tried to remain as neutral as possible in compiling and presenting the more current data with minimal commentary, permitting the reader free reign to extract the knowledge most helpful to their own investigations.
Collapse
Affiliation(s)
- Philip Chiu-Tsun Tang
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Yam-Fung Ng
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China; State Key Laboratory of Chinese Medicine and Molecular Pharmacology, Shenzhen, China
| | - Susan Ho
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Michael Gyda
- Life Sciences Multimedia Productions, Drexel Hill, PA, USA.
| | - Shun-Wan Chan
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China; State Key Laboratory of Chinese Medicine and Molecular Pharmacology, Shenzhen, China; Food Safety and Technology Research Centre, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China.
| |
Collapse
|
19
|
Schuster S, Penke M, Gorski T, Petzold-Quinque S, Damm G, Gebhardt R, Kiess W, Garten A. Resveratrol differentially regulates NAMPT and SIRT1 in Hepatocarcinoma cells and primary human hepatocytes. PLoS One 2014; 9:e91045. [PMID: 24603648 PMCID: PMC3946349 DOI: 10.1371/journal.pone.0091045] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 02/09/2014] [Indexed: 02/08/2023] Open
Abstract
Resveratrol is reported to possess chemotherapeutic properties in several cancers. In this study, we wanted to investigate the molecular mechanisms of resveratrol-induced cell cycle arrest and apoptosis as well as the impact of resveratrol on NAMPT and SIRT1 protein function and asked whether there are differences in hepatocarcinoma cells (HepG2, Hep3B cells) and non-cancerous primary human hepatocytes. We found a lower basal NAMPT mRNA and protein expression in hepatocarcinoma cells compared to primary hepatocytes. In contrast, SIRT1 was significantly higher expressed in hepatocarcinoma cells than in primary hepatocytes. Resveratrol induced cell cycle arrest in the S- and G2/M- phase and apoptosis was mediated by activation of p53 and caspase-3 in HepG2 cells. In contrast to primary hepatocytes, resveratrol treated HepG2 cells showed a reduction of NAMPT enzymatic activity and increased p53 acetylation (K382). Resveratrol induced NAMPT release from HepG2 cells which was associated with increased NAMPT mRNA expression. This effect was absent in primary hepatocytes where resveratrol was shown to function as NAMPT and SIRT1 activator. SIRT1 inhibition by EX527 resembled resveratrol effects on HepG2 cells. Furthermore, a SIRT1 overexpression significantly decreased both p53 hyperacetylation and resveratrol-induced NAMPT release as well as S-phase arrest in HepG2 cells. We could show that NAMPT and SIRT1 are differentially regulated by resveratrol in hepatocarcinoma cells and primary hepatocytes and that resveratrol did not act as a SIRT1 activator in hepatocarcinoma cells.
Collapse
Affiliation(s)
- Susanne Schuster
- Center for Pediatric Research Leipzig, University Hospital for Children and Adolescents, Faculty of Medicine, University of Leipzig, Leipzig, Germany
- * E-mail:
| | - Melanie Penke
- Center for Pediatric Research Leipzig, University Hospital for Children and Adolescents, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Theresa Gorski
- Center for Pediatric Research Leipzig, University Hospital for Children and Adolescents, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Stefanie Petzold-Quinque
- Center for Pediatric Research Leipzig, University Hospital for Children and Adolescents, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Georg Damm
- Department of General-, Visceral- and Transplantation Surgery, Charité University Medicine Berlin, Berlin, Germany
| | - Rolf Gebhardt
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Wieland Kiess
- Center for Pediatric Research Leipzig, University Hospital for Children and Adolescents, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Antje Garten
- Center for Pediatric Research Leipzig, University Hospital for Children and Adolescents, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| |
Collapse
|
20
|
Yang Q, Wang B, Zang W, Wang X, Liu Z, Li W, Jia J. Resveratrol inhibits the growth of gastric cancer by inducing G1 phase arrest and senescence in a Sirt1-dependent manner. PLoS One 2013; 8:e70627. [PMID: 24278101 PMCID: PMC3836800 DOI: 10.1371/journal.pone.0070627] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 06/19/2013] [Indexed: 01/07/2023] Open
Abstract
Resveratrol, a naturally occurring polyphenolic compound, has been reported to exert anticancer activity by affecting diverse molecular targets. In this study, we examined the effects and the underlying mechanisms of resveratrol on gastric cancer. We found that resveratrol inhibited the proliferation of gastric cancer cells in a dose-dependent manner. At the concentration of 25 and 50 µM, resveratrol inhibited the cell viability and diminished the clonogenic potential of gastric cancer cells. Resveratrol treatment arrested gastric cancer cells in the G1 phase and led to senescence instead of apoptosis. Regulators of the cell cycle and senescence pathways, including cyclin D1, cyclin-dependent kinase (CDK4 and 6), p21 and p16, were dysregulated by resveratrol treatment. The inhibitory effects of resveratrol on gastric cancer were also verified in vivo using a nude mice xenograft model. Resveratrol (40 mg/kg/d) exerted inhibitory activities on gastric cancer development and significantly decreased the fractions of Ki67-positive cells in the tumor specimens from the nude mice. After resveratrol treatment, the induction of senescence and the changes in the expression of the regulators involved in the cell cycle and senescence pathways were similar to what we observed in vitro. However, the depletion of Sirtuin (Sirt)1 reversed the above-described effects of resveratrol both in vitro and in vivo. Our data suggest that resveratrol inhibits gastric cancer in a Sirt1-dependent manner and provide detailed evidence for the possibility of applying resveratrol in gastric cancer prevention and therapy.
Collapse
Affiliation(s)
- Qing Yang
- Institute of Pathogen Biology, Shandong University School of Medicine, Jinan, Shandong Province, China
| | - Bo Wang
- Department of Traditional Chinese Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Wen Zang
- Institute of Pathogen Biology, Shandong University School of Medicine, Jinan, Shandong Province, China
| | - Xuping Wang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Zhifang Liu
- Department of Biochemistry, Shandong University School of Medicine, Jinan, Shandong Province, China
| | - Wenjuan Li
- Institute of Pathogen Biology, Shandong University School of Medicine, Jinan, Shandong Province, China
| | - Jihui Jia
- Institute of Pathogen Biology, Shandong University School of Medicine, Jinan, Shandong Province, China
- * E-mail:
| |
Collapse
|
21
|
Woo SR, Byun JG, Kim YH, Park ER, Joo HY, Yun M, Shin HJ, Kim SH, Shen YN, Park JE, Park GH, Lee KH. SIRT1 suppresses cellular accumulation of β-TrCP E3 ligase via protein degradation. Biochem Biophys Res Commun 2013; 441:831-7. [PMID: 24211209 DOI: 10.1016/j.bbrc.2013.10.146] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 10/28/2013] [Indexed: 10/26/2022]
Abstract
β-Transducin repeat-containing protein (β-TrCP), an E3 ligase, promotes the degradation of substrate proteins in response to various stimuli. Even though several β-TrCP substrates have been identified to date, limited information of its upstream regulators is available. Here, we showed that SIRT1 suppresses β-TrCP protein synthesis via post-translational degradation. SIRT1 depletion led to a significant increase in the β-TrCP accumulation without affecting the mRNA level. Consistently, β-TrCP protein accumulation induced by resveratrol was further enhanced upon SIRT1 depletion. Rescue of SIRT1 reversed the effect of resveratrol, leading to reduced β-TrCP protein levels. Proteasomal inhibition led to recovery of β-TrCP in cells with SIRT1 overexpression. Notably, the recovered β-TrCP colocalized mostly with SIRT1. Thus, SIRT1 acts as a negative regulator of β-TrCP synthesis via promoting protein degradation.
Collapse
Affiliation(s)
- Seon Rang Woo
- Division of Radiation Cancer Research, Korea Institute of Radiological & Medical Sciences, Seoul 139-706, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Sun Z, Shi S, Li H, Shu XH, Chen XY, Kong QY, Liu J. Evaluation of resveratrol sensitivities and metabolic patterns in human and rat glioblastoma cells. Cancer Chemother Pharmacol 2013; 72:965-73. [PMID: 23989725 DOI: 10.1007/s00280-013-2274-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 08/15/2013] [Indexed: 02/05/2023]
Abstract
PURPOSE To further elucidate the correlation of resveratrol sensitivities with biotransformation activities of human and rat glioblastoma cells for personalized anti-glioblastoma therapy. METHODS Resveratrol sensitivity of human U251 and rat RG2 and C6 glioblastoma cells was evaluated by 3-[4,5-dimethylthiazol-2-yl]-2, 5-diphenyltetrazolium bromide/MTT, flow cytometry, and TUNEL assays. The metabolic patterns of those cell lines were analyzed by high-performance liquid chromatography/HPLC coupled with tandem mass spectrum/MS/MS, and high-resolution mass spectrometry/HRMS. Immunocytochemical staining and Western blotting were employed to check resveratrol metabolic enzyme expression. RESULTS Both rat RG2 and C6 and human U251 glioblastoma cells are sensitive to 100 μM resveratrol in terms of growth arrest and increased apoptotic fraction. The main resveratrol metabolite in U251 cells is monosulfate biotransformed by sulfotransferases/SULTs and in RG2 and C6 cells is monoglucuronide generated by UDP-glucuronosyltransferase/UGT. Both metabolites show lesser therapeutic efficacy. Although brain-associated UGTs (UGT1A6, 2B7, and 8) and SULTs (SULT1A1, 1C2, and 4A1) are expressed in rat and human glioma cells, the overall level of UGTs is predominant in the rat and SULTs in human glioblastoma cells. In similar to SULT expression pattern, UGT1A6, 2B7, and 8 are frequently downregulated (84.6 %, 82/97; 90.7 %, 88/97; 80.4 %, 78/97) in human glioblastoma tissues. CONCLUSION Our results suggest (1) the decreased resveratrol biotransforming activity in rat and human resveratrol-sensitive glioblastoma cells; (2) the discrepant resveratrol metabolic patterns between human and rat glioblastoma cells; (3) the more powerful anti-glioblastoma efficacy of trans-resveratrol rather than resveratrol monoglucuronide or monosulfate; and (4) the value of RG2 and C6 cells in establishing resveratrol-based rat in vivo therapeutic model.
Collapse
Affiliation(s)
- Zheng Sun
- Liaoning Laboratory of Cancer Genomics, Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian, 116044, China
| | | | | | | | | | | | | |
Collapse
|
23
|
Stakleff KS, Sloan T, Blanco D, Marcanthony S, Booth TD, Bishayee A. Resveratrol exerts differential effects in vitro and in vivo against ovarian cancer cells. Asian Pac J Cancer Prev 2013; 13:1333-40. [PMID: 22799328 DOI: 10.7314/apjcp.2012.13.4.1333] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Epithelial ovarian cancer represents the most lethal gynecological cancer, and the high mortality rate makes this malignancy a major health concern. Poor prognosis results from an inability to detect ovarian cancers at an early, curable stage, as well as from the lack of an effective therapy. Thus, effective and novel strategies for prevention and treatment with non-toxic agents merit serious consideration. Resveratrol, obtained from grapes, berries, peanuts and red wine, has been shown to have a potent growth-inhibitory effect against various human cancer cells as well as in in vivo preclinical cancer models. The objective here was to evaluate potential antitumor effects of resveratrol in both in vitro and in vivo NuTu-19 ovarian cancer models. In vitro an invasion assay was performed. After 48 h, the numbers of viable cells that invaded the extracellular matrix layer were reduced by 94% with resveratrol in comparison to control. For the in vivo anti-tumor assessment, 10 rats were injected with NuTu-19 cells into the ovarian bursa. Thereafter, half were provided with a diet mixed with a dose of 100 mg resveratrol/kg body weight/day for 28 days. Following sacrifice, anticancer effects were assessed by histological evaluation of ovarian as well as surrounding tissues, and immunohistochemical detection of cell proliferation and apoptosis, but there were no observable differences between the control and resveratrol-treated groups for any of the biological endpoints. While resveratrol is effective in suppressing the in vitro cellular invasion of NuTu-19 ovarian cancer cells, these effects do not appear to impact on in vivo NuTu-19 ovarian cancers in rats.
Collapse
|
24
|
Quoc Trung L, Espinoza JL, Takami A, Nakao S. Resveratrol induces cell cycle arrest and apoptosis in malignant NK cells via JAK2/STAT3 pathway inhibition. PLoS One 2013; 8:e55183. [PMID: 23372833 PMCID: PMC3555980 DOI: 10.1371/journal.pone.0055183] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 12/19/2012] [Indexed: 02/06/2023] Open
Abstract
Natural killer (NK) cell malignancies, particularly aggressive NK cell leukaemias and lymphomas, have poor prognoses. Although recent regimens with L-asparaginase substantially improved outcomes, novel therapeutic approaches are still needed to enhance clinical response. Resveratrol, a naturally occurring polyphenol, has been extensively studied for its anti-inflammatory, cardioprotective and anti-cancer activities. In this study, we investigated the potential anti-tumour activities of resveratrol against the NK cell lines KHYG-1, NKL, NK-92 and NK-YS. Resveratrol induced robust G0/G1 cell cycle arrest, significantly suppressed cell proliferation and induced apoptosis in a dose- and time-dependent manner for all four cell lines. In addition, resveratrol suppressed constitutively active STAT3 in all the cell lines and inhibited JAK2 phosphorylation but had no effect on other upstream mediators of STAT3 activation, such as PTEN, TYK2, and JAK1. Resveratrol also induced downregulation of the anti-apoptotic proteins MCL1 and survivin, two downstream effectors of the STAT3 pathway. Finally, resveratrol induced synergistic effect on the apoptotic and antiproliferative activities of L-asparaginase against KHYG-1, NKL and NK-92 cells. These results suggest that resveratrol may have therapeutic potential against NK cell malignancies. Furthermore, our finding that resveratrol is a bonafide JAK2 inhibitor extends its potential benefits to other diseases with dysregulated JAK2 signaling.
Collapse
Affiliation(s)
- Ly Quoc Trung
- Cellular Transplantation Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - J. Luis Espinoza
- Cellular Transplantation Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
- * E-mail:
| | - Akiyoshi Takami
- Department of Haematology and Oncology, Kanazawa University Hospital, Ishikawa, Japan
| | - Shinji Nakao
- Cellular Transplantation Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| |
Collapse
|
25
|
Wintzell M, Löfstedt L, Johansson J, Pedersen AB, Fuxe J, Shoshan M. Repeated cisplatin treatment can lead to a multiresistant tumor cell population with stem cell features and sensitivity to 3-bromopyruvate. Cancer Biol Ther 2012; 13:1454-62. [PMID: 22954696 DOI: 10.4161/cbt.22007] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cisplatin is used in treatment of several types of cancer, including epithelial ovarian carcinoma (EOC). In order to mimic clinical treatment and to investigate longterm effects of cisplatin in surviving cancer cells, two EOC cell lines were repeatedly treated with low doses. In the SKOV-3 cell line originating from malignant ascites, but not in A2780 cells from a primary tumor, this led to emergence of a stable population (SKOV-3-R) which in the absence of cisplatin showed increased motility, epithelial-mesenchymal transition (EMT) and expression of cancer stem cell markers CD117, CD44 and ALDH1. Accordingly, the cells formed self-renewing spheres in serum-free stem cell medium. Despite upregulation of mitochondrial mass and cytochrome c, and no upregulation of Bcl-2/Bcl-xL, SKOV-3-R were multiresistant to antineoplastic drugs. Cancer stem cells, or tumor-initiating cells (TICs) are highly chemoresistant and are believed to cause relapse into disseminated and resistant EOC. Our second aim was therefore to target resistance in these TIC-like cells. Resistance could be correlated with upregulation of hexokinase-II and VDAC, which are known to form a survival-promoting mitochondrial complex. The cells were thus sensitive to 3-bromopyruvate, which dissociates hexokinase-II from this complex, and were particularly sensitive to combination treatment with cisplatin at doses down to 0.1 x IC 50. 3-bromopyruvate might thus be of use in targeting the especially aggressive TIC populations.
Collapse
Affiliation(s)
- My Wintzell
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
26
|
Price NL, Gomes AP, Ling AJ, Duarte FV, Martin-Montalvo A, North BJ, Agarwal B, Ye L, Ramadori G, Teodoro JS, Hubbard BP, Varela AT, Davis JG, Varamini B, Hafner A, Moaddel R, Rolo AP, Coppari R, Palmeira CM, de Cabo R, Baur JA, Sinclair DA. SIRT1 is required for AMPK activation and the beneficial effects of resveratrol on mitochondrial function. Cell Metab 2012; 15:675-90. [PMID: 22560220 PMCID: PMC3545644 DOI: 10.1016/j.cmet.2012.04.003] [Citation(s) in RCA: 1155] [Impact Index Per Article: 96.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 02/14/2012] [Accepted: 04/06/2012] [Indexed: 02/06/2023]
Abstract
Resveratrol induces mitochondrial biogenesis and protects against metabolic decline, but whether SIRT1 mediates these benefits is the subject of debate. To circumvent the developmental defects of germline SIRT1 knockouts, we have developed an inducible system that permits whole-body deletion of SIRT1 in adult mice. Mice treated with a moderate dose of resveratrol showed increased mitochondrial biogenesis and function, AMPK activation, and increased NAD(+) levels in skeletal muscle, whereas SIRT1 knockouts displayed none of these benefits. A mouse overexpressing SIRT1 mimicked these effects. A high dose of resveratrol activated AMPK in a SIRT1-independent manner, demonstrating that resveratrol dosage is a critical factor. Importantly, at both doses of resveratrol no improvements in mitochondrial function were observed in animals lacking SIRT1. Together these data indicate that SIRT1 plays an essential role in the ability of moderate doses of resveratrol to stimulate AMPK and improve mitochondrial function both in vitro and in vivo.
Collapse
Affiliation(s)
- Nathan L. Price
- Glenn Labs for the Biological Mechanisms of Aging, Harvard Medical School, Boston, MA, 02115
| | - Ana P. Gomes
- Glenn Labs for the Biological Mechanisms of Aging, Harvard Medical School, Boston, MA, 02115
- Center for Neurosciences and Cell Biology, 3004-517 Coimbra, Portugal
| | - Alvin J.Y. Ling
- Glenn Labs for the Biological Mechanisms of Aging, Harvard Medical School, Boston, MA, 02115
| | - Filipe V. Duarte
- Center for Neurosciences and Cell Biology, 3004-517 Coimbra, Portugal
| | - Alejandro Martin-Montalvo
- Laboratory of Experimental Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Brian J. North
- Glenn Labs for the Biological Mechanisms of Aging, Harvard Medical School, Boston, MA, 02115
| | - Beamon Agarwal
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Lan Ye
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Giorgio Ramadori
- Department of Internal Medicine, Division of Hypothalamic Research, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joao S. Teodoro
- Center for Neurosciences and Cell Biology, 3004-517 Coimbra, Portugal
| | - Basil P. Hubbard
- Glenn Labs for the Biological Mechanisms of Aging, Harvard Medical School, Boston, MA, 02115
| | - Ana T. Varela
- Center for Neurosciences and Cell Biology, 3004-517 Coimbra, Portugal
| | - James G. Davis
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Behzad Varamini
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Angela Hafner
- Glenn Labs for the Biological Mechanisms of Aging, Harvard Medical School, Boston, MA, 02115
| | - Ruin Moaddel
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Anabela P. Rolo
- Center for Neurosciences and Cell Biology, 3004-517 Coimbra, Portugal
- Department of Biology, University of Aveiro, 3810-193, Aveiro Portugal
| | - Roberto Coppari
- Department of Internal Medicine, Division of Hypothalamic Research, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Dipartimento di Medicina Sperimentale e Clinica, Universita’ Politecnica delle Marche, Ancona 60020, Italy
| | - Carlos M. Palmeira
- Center for Neurosciences and Cell Biology, 3004-517 Coimbra, Portugal
- Department of Life Sciences, Faculty of Science and Technology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Rafael de Cabo
- Laboratory of Experimental Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Joseph A. Baur
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - David A. Sinclair
- Glenn Labs for the Biological Mechanisms of Aging, Harvard Medical School, Boston, MA, 02115
- Corresponding author: David A. Sinclair ()
| |
Collapse
|
27
|
Stressing the ubiquitin-proteasome system without 20S proteolytic inhibition selectively kills cervical cancer cells. PLoS One 2011; 6:e23888. [PMID: 21909374 PMCID: PMC3166081 DOI: 10.1371/journal.pone.0023888] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 07/29/2011] [Indexed: 12/22/2022] Open
Abstract
Cervical cancer cells exhibit an increased requirement for ubiquitin-dependent protein degradation associated with an elevated metabolic turnover rate, and for specific signaling pathways, notably HPV E6-targeted degradation of p53 and PDZ proteins. Natural compounds with antioxidant properties including flavonoids and triterpenoids hold promise as anticancer agents by interfering with ubiquitin-dependent protein degradation. An increasing body of evidence indicates that their α-β unsaturated carbonyl system is the molecular determinant for inhibition of ubiquitin-mediated protein degradation up-stream of the catalytic sites of the 20S proteasome. Herein we report the identification and characterization of a new class of chalcone-based, potent and cell permeable chemical inhibitors of ubiquitin-dependent protein degradation, and a lead compound RAMB1. RAMB1 inhibits ubiquitin-dependent protein degradation without compromising the catalytic activities of the 20S proteasome, a mechanism distinct from that of Bortezomib. Treatment of cervical cancer cells with RAMB1 triggers unfolded protein responses, including aggresome formation and Hsp90 stabilization, and increases p53 steady state levels. RAMB1 treatment results in activation of lysosomal-dependent degradation pathways as a mechanism to compensate for increasing levels of poly-ubiquitin enriched toxic aggregates. Importantly, RAMB1 synergistically triggers cell death of cervical cancer cells when combined with the lysosome inhibitor Chloroquine.
Collapse
|
28
|
Closer association of mitochondria with lipid droplets in hepatocytes and activation of Kupffer cells in resveratrol-treated senescence-accelerated mice. Histochem Cell Biol 2011; 136:475-89. [PMID: 21818579 DOI: 10.1007/s00418-011-0847-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2011] [Indexed: 12/13/2022]
Abstract
Resveratrol has been extensively investigated because of its beneficial effects in delaying age-related diseases, thus extending the lifespan, possibly by mimicking calorie restriction. For this study, cell biological techniques were used to examine how resveratrol influenced hepatocytes in a senescence-accelerated mouse P10 (SAMP10), treated from 35 to 55 weeks of age, with special emphasis on the relationship between mitochondria and lipid droplets. Survival ratio, body weight and food intake of SAMP10 did not differ significantly between the control and resveratrol-treated groups. Compared with the control, the treated livers were altered significantly, as follows. Lipid droplets were reduced and mitochondria were increased in number in hepatocytes. Phosphorylation of acetyl-CoA carboxylase and the expression of both the mitochondrial ATP synthase β subunit and Mn superoxide dismutase (SOD2) were increased. Mitochondria, expressing more SOD2, were more tightly associated with lipid droplets, suggesting the enhancement of lipolysis through the activation of mitochondrial functions. Cathepsin D expression was less in hepatocytes but enhanced in Kupffer cells, which were increased in number and size with more numerous lysosome-related profiles. Together, resveratrol may activate mitochondria resulting in consuming lipids, and may also activate Kupffer cells by which a beneficial milieu for hepatocytes may be created. Both might be related to improvement in the functioning of the liver, which is the organ that is central to metabolic regulation.
Collapse
|