1
|
Schol P, van Elsas MJ, Middelburg J, Nijen Twilhaar MK, van Hall T, van der Sluis TC, van der Burg SH. Myeloid effector cells in cancer. Cancer Cell 2024; 42:1997-2014. [PMID: 39658540 DOI: 10.1016/j.ccell.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/21/2024] [Accepted: 11/06/2024] [Indexed: 12/12/2024]
Abstract
The role of myeloid cells in tumor immunity is multifaceted. While dendritic cells support T cell-mediated tumor control, the highly heterogenous populations of macrophages, neutrophils, and immature myeloid cells were generally considered immunosuppressive. This view has led to effective therapies reinvigorating tumor-reactive T cells; however, targeting the immunosuppressive effects of macrophages and neutrophils to boost the cancer immunity cycle was clinically less successful. Recent studies interrogating the role of immune cells in the context of successful immunotherapy affirm the key role of T cells, but simultaneously challenge the idea that the cytotoxic function of T cells is the main contributor to therapy-driven tumor regression. Rather, therapy-activated intra-tumoral T cells recruit and activate or reprogram several myeloid effector cell types, the presence of which is necessary for tumor rejection. Here, we reappreciate the key role of myeloid effector cells in tumor rejection as this may help to shape future successful immunotherapies.
Collapse
Affiliation(s)
- Pieter Schol
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Marit J van Elsas
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Jim Middelburg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Maarten K Nijen Twilhaar
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Thorbald van Hall
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Tetje C van der Sluis
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
2
|
Shin Y. Histone Tail Cleavage as a Mechanism for Epigenetic Regulation. Int J Mol Sci 2024; 25:10789. [PMID: 39409117 PMCID: PMC11477362 DOI: 10.3390/ijms251910789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
Histones are essential for DNA packaging and undergo post-translational modifications that significantly influence gene regulation. Among these modifications, histone tail cleavage has recently garnered attention despite being less explored. Cleavage by various proteases impacts processes such as stem cell differentiation, aging, infection, and inflammation, though the mechanisms remain unclear. This review delves into recent insights on histone proteolytic cleavage and its epigenetic significance, highlighting how chromatin, which serves as a dynamic scaffold, responds to signals through histone modification, replacement, and ATP-dependent remodeling. Specifically, histone tail cleavage is linked to critical cellular processes such as granulocyte differentiation, viral infection, aging, yeast sporulation, and cancer development. Although the exact mechanisms connecting histone cleavage to gene expression are still emerging, it is clear that this process represents a novel epigenetic transcriptional mechanism intertwined with chromatin dynamics. This review explores known histone tail cleavage events, the proteolytic enzymes involved, their impact on gene expression, and future research directions in this evolving field.
Collapse
Affiliation(s)
- Yonghwan Shin
- Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
3
|
Yang T, Peng J, Zhang Z, Chen Y, Liu Z, Jiang L, Jin L, Han M, Su B, Li Y. Emerging therapeutic strategies targeting extracellular histones for critical and inflammatory diseases: an updated narrative review. Front Immunol 2024; 15:1438984. [PMID: 39206200 PMCID: PMC11349558 DOI: 10.3389/fimmu.2024.1438984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Extracellular histones are crucial damage-associated molecular patterns involved in the development and progression of multiple critical and inflammatory diseases, such as sepsis, pancreatitis, trauma, acute liver failure, acute respiratory distress syndrome, vasculitis and arthritis. During the past decade, the physiopathologic mechanisms of histone-mediated hyperinflammation, endothelial dysfunction, coagulation activation, neuroimmune injury and organ dysfunction in diseases have been systematically elucidated. Emerging preclinical evidence further shows that anti-histone strategies with either their neutralizers (heparin, heparinoids, nature plasma proteins, small anion molecules and nanomedicines, etc.) or extracorporeal blood purification techniques can significantly alleviate histone-induced deleterious effects, and thus improve the outcomes of histone-related critical and inflammatory animal models. However, a systemic evaluation of the efficacy and safety of these histone-targeting therapeutic strategies is currently lacking. In this review, we first update our latest understanding of the underlying molecular mechanisms of histone-induced hyperinflammation, endothelial dysfunction, coagulopathy, and organ dysfunction. Then, we summarize the latest advances in histone-targeting therapy strategies with heparin, anti-histone antibodies, histone-binding proteins or molecules, and histone-affinity hemoadsorption in pre-clinical studies. Finally, challenges and future perspectives for improving the clinical translation of histone-targeting therapeutic strategies are also discussed to promote better management of patients with histone-related diseases.
Collapse
Affiliation(s)
- Tinghang Yang
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Jing Peng
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Zhuyun Zhang
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Yu Chen
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu, China
| | - Zhihui Liu
- Department of Rheumatology and Immunology, West China Hospital of Sichuan University, Chengdu, China
| | - Luojia Jiang
- Jiujiang City Key Laboratory of Cell Therapy, Department of Nephrology, Jiujiang No. 1 People’s Hospital, Jiujiang, China
| | - Lunqiang Jin
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Mei Han
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Baihai Su
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
- Med+ Biomaterial Institute of West China Hospital/West China School of Medicine, Sichuan University, Chengdu, China
- Med-X Center for Materials, Sichuan University, Chengdu, China
| | - Yupei Li
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Gong Y, Wei C, Cheng L, Ma F, Lu S, Peng Q, Liu L, Wang Y. Tracking the Dynamic Histone Methylation of H3K27 in Live Cancer Cells. ACS Sens 2021; 6:4369-4378. [PMID: 34878766 PMCID: PMC9013700 DOI: 10.1021/acssensors.1c01670] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Histone methylations play a crucial role in chromatin remodeling and genome regulations. However, there is a lack of tools to visualize these histone modifications with high spatiotemporal resolutions in live cells. We have developed a biosensor based on fluorescence resonance energy transfer (FRET) and incorporated it into nucleosomes, capable of monitoring the trimethylation of H3K27 (H3K27me3) in live cells. We also revealed that the performance of the FRET biosensor can be significantly improved by adjusting the linkers within the biosensor. An improved biosensor enables the live-cell imaging of different histone methylation status, induced by the suppressive H3.3K27M or existing in breast cancer cells with varying genetic backgrounds. We have further applied the biosensor to reveal the dynamic coupling between H3K27me3 changes and caspase activity representing the initiation of apoptosis in cancer cells by imaging both H3K27me3 and caspase activity simultaneously in the same live cells. Thus, this new FRET biosensor can provide a powerful tool to visualize the epigenetic regulation in live cells with high spatial temporal resolutions.
Collapse
Affiliation(s)
- Ya Gong
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California 92093-0435, United States
| | - Chujun Wei
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California 92093-0435, United States
| | - Leonardo Cheng
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California 92093-0435, United States
| | - Fengyi Ma
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California 92093-0435, United States
| | - Shaoying Lu
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California 92093-0435, United States
| | - Qin Peng
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California 92093-0435, United States
| | - Longwei Liu
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California 92093-0435, United States
| | - Yingxiao Wang
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California 92093-0435, United States
| |
Collapse
|
5
|
Effect of histone H4 tail on nucleosome stability and internucleosomal interactions. Sci Rep 2021; 11:24086. [PMID: 34916563 PMCID: PMC8677776 DOI: 10.1038/s41598-021-03561-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/02/2021] [Indexed: 11/28/2022] Open
Abstract
Chromatin structure is dictated by nucleosome assembly and internucleosomal interactions. The tight wrapping of nucleosomes inhibits gene expression, but modifications to histone tails modulate chromatin structure, allowing for proper genetic function. The histone H4 tail is thought to play a large role in regulating chromatin structure. Here we investigated the structure of nucleosomes assembled with a tail-truncated H4 histone using Atomic Force Microscopy. We assembled tail-truncated H4 nucleosomes on DNA templates allowing for the assembly of mononucleosomes or dinucleosomes. Mononucleosomes assembled on nonspecific DNA led to decreased DNA wrapping efficiency. This effect is less pronounced for nucleosomes assembled on positioning motifs. Dinucleosome studies resulted in the discovery of two effects- truncation of the H4 tail does not diminish the preferential positioning observed in full-length nucleosomes, and internucleosomal interaction eliminates the DNA unwrapping effect. These findings provide insight on the role of histone H4 in chromatin structure and stability.
Collapse
|
6
|
Involvement of p53-dependent apoptosis signal in antitumor effect of Colchicine on human papilloma virus (HPV)-positive human cervical cancer cells. Biosci Rep 2021; 40:222342. [PMID: 32163135 PMCID: PMC7098170 DOI: 10.1042/bsr20194065] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/28/2020] [Accepted: 03/09/2020] [Indexed: 12/15/2022] Open
Abstract
Colchicine, a plant-derived alkaloid with relatively low toxicity on normal human epidermal keratinocytes (HEKn), has selective inhibitory effect on the growth of CaSki (HPV16-positive) and HeLa (HPV18-positive) human cervical cancer cell lines via the induction of apoptosis. Colchicine (2.5, 5.0 and 10.0 ng/ml) significantly reduced the expression of human papilloma virus (HPV) 16 E6/E7 mRNA and protein in CaSki and HeLa cells. Moreover, reduced expression of E6 and E7 induced by Colchicine resulted in the up-regulation of tumor suppressor proteins, p53 and Rb, as well as down-regulation of phospho Rb (pRb) protein. In addition, Bax, cytosolic cytochrome c and cleaved caspase-3 protein were increased while Bcl-2 protein was decreased significantly by 48 h of Colchicine treatment. These results implied that Colchicine could be explored as a potent candidate agent for the treatment and prevention of HPV-associated cervical cancer without deleterious effects.
Collapse
|
7
|
Florean C, Song S, Dicato M, Diederich M. Redox biology of regulated cell death in cancer: A focus on necroptosis and ferroptosis. Free Radic Biol Med 2019; 134:177-189. [PMID: 30639617 DOI: 10.1016/j.freeradbiomed.2019.01.008] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/23/2018] [Accepted: 01/06/2019] [Indexed: 12/20/2022]
Abstract
Redox changes and generation of reactive oxygen species (ROS) are part of normal cell metabolism. While low ROS levels are implicated in cellular signaling pathways necessary for survival, higher levels play major roles in cancer development as well as cell death signaling and execution. A role for redox changes in apoptosis has been long established; however, several new modalities of regulated cell death have been brought to light, for which the importance of ROS production as well as ROS source and targets are being actively investigated. In this review, we summarize recent findings on the role of ROS and redox changes in the activation and execution of two major forms of regulated cell death, necroptosis and ferroptosis. We also discuss the potential of using modulators of these two forms of cell death to exacerbate ROS as a promising anticancer therapy.
Collapse
Affiliation(s)
- Cristina Florean
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer (LBMCC), Hôpital Kirchberg, L-2540 Luxembourg, Luxembourg
| | - Sungmi Song
- Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Mario Dicato
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer (LBMCC), Hôpital Kirchberg, L-2540 Luxembourg, Luxembourg
| | - Marc Diederich
- Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| |
Collapse
|
8
|
Abstract
Chromatin is an intelligent building block that can express either external or internal needs through structural changes. To date, three methods to change chromatin structure and regulate gene expression have been well-documented: histone modification, histone exchange, and ATP-dependent chromatin remodeling. Recently, a growing body of literature has suggested that histone tail cleavage is related to various cellular processes including stem cell differentiation, osteoclast differentiation, granulocyte differentiation, mammary gland differentiation, viral infection, aging, and yeast sporulation. Although the underlying mechanisms suggesting how histone cleavage affects gene expression in view of chromatin structure are only beginning to be understood, it is clear that this process is a novel transcriptional epigenetic mechanism involving chromatin dynamics. In this review, we describe the functional properties of the known histone tail cleavage with its proteolytic enzymes, discuss how histone cleavage impacts gene expression, and present future directions for this area of study.
Collapse
Affiliation(s)
- Sun-Ju Yi
- School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju 28644, Korea
| | - Kyunghwan Kim
- School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
9
|
Kim MY, Bo HH, Choi EO, Kwon DH, Kim HJ, Ahn KI, Ji SY, Jeong JW, Park SH, Hong SH, Kim GY, Park C, Kim HS, Moon SK, Yun SJ, Kim WJ, Choi YH. Induction of Apoptosis by Citrus unshiu Peel in Human Breast Cancer MCF-7 Cells: Involvement of ROS-Dependent Activation of AMPK. Biol Pharm Bull 2018; 41:713-721. [PMID: 29709909 DOI: 10.1248/bpb.b17-00898] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The fruit of Citrus unshiu MARKOVICH used for various purposes in traditional medicine has various pharmacological properties including antioxidant, anti-inflammatory, and antibacterial effects. Recently, the possibility of anti-cancer activity of the extracts or components of this fruit has been reported; however, the exact mechanism has not yet been fully understood. In this study, we evaluated the anti-proliferative effect of water extract of C. unshiu peel (WECU) on human breast cancer MCF-7 cells and investigated the underlying mechanism. Our results showed that reduction of MCF-7 cell survival by WECU was associated with the induction of apoptosis. WECU-induced apoptotic cell death was related to the activation of caspase-8 and -9, representative initiate caspases of extrinsic and intrinsic apoptosis pathways, respectively, and increase in the Bax : Bcl-2 ratio accompanied by cleavage of poly(ADP-ribose) polymerase (PARP). WECU also increased the mitochondrial dysfunction and cytosolic release of cytochrome c. In addition, AMP-activated protein kinase (AMPK) and its downstream target molecule, acetyl-CoA carboxylase, were activated in a concentration-dependent manner in WECU-treated cells. In contrast, compound C, an AMPK inhibitor, significantly inhibited WECU-induced apoptosis, while inhibiting increased expression of Bax and decreased expression of Bcl-2 by WECU and inhibition of WECU-induced PARP degradation. Furthermore, WECU provoked the production of reactive oxygen species (ROS); however, the activation of AMKP and apoptosis by WECU were prevented, when the ROS production was blocked by antioxidant N-acetyl cysteine. Therefore, our data indicate that WECU suppresses MCF-7 cell proliferation by activating the intrinsic and extrinsic apoptosis pathways through ROS-dependent AMPK pathway activation.
Collapse
Affiliation(s)
- Min Yeong Kim
- Department of Biochemistry, Dongeui University College of Korean Medicine.,Anti-Aging Research Center, Dongeui University
| | - Hyun Hwang Bo
- Department of Biochemistry, Dongeui University College of Korean Medicine.,Anti-Aging Research Center, Dongeui University
| | - Eun Ok Choi
- Department of Biochemistry, Dongeui University College of Korean Medicine.,Anti-Aging Research Center, Dongeui University
| | - Da He Kwon
- Department of Biochemistry, Dongeui University College of Korean Medicine.,Anti-Aging Research Center, Dongeui University
| | - Hong Jae Kim
- Department of Biochemistry, Dongeui University College of Korean Medicine.,Anti-Aging Research Center, Dongeui University
| | - Kyu Im Ahn
- Department of Biochemistry, Dongeui University College of Korean Medicine.,Anti-Aging Research Center, Dongeui University
| | - Seon Yeong Ji
- Department of Biochemistry, Dongeui University College of Korean Medicine.,Anti-Aging Research Center, Dongeui University
| | - Jin-Woo Jeong
- Department of Biochemistry, Dongeui University College of Korean Medicine.,Anti-Aging Research Center, Dongeui University
| | - Shin-Hyung Park
- Department of Pathology, Dongeui University College of Korean Medicine
| | - Su-Hyun Hong
- Department of Biochemistry, Dongeui University College of Korean Medicine.,Anti-Aging Research Center, Dongeui University
| | - Gi-Young Kim
- Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National University
| | - Cheol Park
- Department of Molecular Biology, College of Natural Sciences, Dongeui University
| | - Heui-Soo Kim
- Department of Biological Sciences, College of Natural Sciences, Pusan National University
| | - Sung-Kwon Moon
- Department of Food and Nutrition, College of Biotechnology & Natural Resource, Chung-Ang University
| | - Seok-Joong Yun
- Personalized Tumor Engineering Research Center, Department of Urology, Chungbuk National University College of Medicine
| | - Wun Jae Kim
- Personalized Tumor Engineering Research Center, Department of Urology, Chungbuk National University College of Medicine
| | - Yung Hyun Choi
- Department of Biochemistry, Dongeui University College of Korean Medicine.,Anti-Aging Research Center, Dongeui University
| |
Collapse
|
10
|
Azad GK, Swagatika S, Kumawat M, Kumawat R, Tomar RS. Modifying Chromatin by Histone Tail Clipping. J Mol Biol 2018; 430:3051-3067. [DOI: 10.1016/j.jmb.2018.07.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/10/2018] [Accepted: 07/10/2018] [Indexed: 12/15/2022]
|
11
|
Kim MY, Choi EO, HwangBo H, Kwon DH, Ahn KI, Kim HJ, Ji SY, Hong SH, Jeong JW, Kim GY, Park C, Choi YH. Reactive oxygen species-dependent apoptosis induction by water extract of Citrus unshiu peel in MDA-MB-231 human breast carcinoma cells. Nutr Res Pract 2018; 12:129-134. [PMID: 29629029 PMCID: PMC5886964 DOI: 10.4162/nrp.2018.12.2.129] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 11/10/2017] [Accepted: 01/29/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND/OBJECTIVES Although several recent studies have reported the anti-cancer effects of extracts or components of Citrus unshiu peel, which has been used for various purposes in traditional medicine, the molecular mechanisms for their effects remain unclear. In the present study, the anti-cancer activity of a water-soluble extract of C. unshiu peel (WECU) in MDA-MB-231 human breast carcinoma cells at the level of apoptosis induction was investigated. MATERIALS/METHODS Cytotoxicity was evaluated using the MTT assay. Apoptosis was detected using DAPI staining and flow cytometry analyses. Mitochondrial membrane potential, reactive oxygen species (ROS) assay, caspase activity and Western blotting were used to confirm the basis of apoptosis. RESULTS The results indicated that WECU-induced apoptosis was related to the activation of caspase-8, and -9, representative initiator caspases of extrinsic and intrinsic apoptosis pathways, respectively, and caspase-3 accompanied by proteolytic degradation of poly(ADP-ribose) polymerase and down-regulation of the inhibitors of apoptosis protein family members. WECU also increased the pro-apoptotic BAX to anti-apoptotic BCL-2 ratio, loss of mitochondrial membrane potential and cytochrome c release from mitochondria to cytoplasm. Furthermore, WECU provoked the generation of ROS, but the reduction of cell viability and induction of apoptosis by WECU were prevented when ROS production was blocked by antioxidant N-acetyl cysteine. CONCLUSIONS These results suggest that WECU suppressed proliferation of MDA-MB-231 cells by activating extrinsic and intrinsic apoptosis pathways in a ROS-dependent manner.
Collapse
Affiliation(s)
- Min Yeong Kim
- Open Laboratory for Muscular and Skeletal Disease, and Department of Biochemistry, Dongeui University College of Korean Medicine, 42 San, Yangjungdong, Busan 47227, Korea.,Anti-Aging Research Center, Dongeui University, Busan 47340, Korea
| | - Eun Ok Choi
- Open Laboratory for Muscular and Skeletal Disease, and Department of Biochemistry, Dongeui University College of Korean Medicine, 42 San, Yangjungdong, Busan 47227, Korea.,Anti-Aging Research Center, Dongeui University, Busan 47340, Korea
| | - Hyun HwangBo
- Open Laboratory for Muscular and Skeletal Disease, and Department of Biochemistry, Dongeui University College of Korean Medicine, 42 San, Yangjungdong, Busan 47227, Korea.,Anti-Aging Research Center, Dongeui University, Busan 47340, Korea
| | - Da He Kwon
- Open Laboratory for Muscular and Skeletal Disease, and Department of Biochemistry, Dongeui University College of Korean Medicine, 42 San, Yangjungdong, Busan 47227, Korea.,Anti-Aging Research Center, Dongeui University, Busan 47340, Korea
| | - Kyu Im Ahn
- Open Laboratory for Muscular and Skeletal Disease, and Department of Biochemistry, Dongeui University College of Korean Medicine, 42 San, Yangjungdong, Busan 47227, Korea.,Anti-Aging Research Center, Dongeui University, Busan 47340, Korea.,Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National University, Jeju 63243, Korea
| | - Hong Jae Kim
- Open Laboratory for Muscular and Skeletal Disease, and Department of Biochemistry, Dongeui University College of Korean Medicine, 42 San, Yangjungdong, Busan 47227, Korea.,Anti-Aging Research Center, Dongeui University, Busan 47340, Korea
| | - Seon Yeong Ji
- Open Laboratory for Muscular and Skeletal Disease, and Department of Biochemistry, Dongeui University College of Korean Medicine, 42 San, Yangjungdong, Busan 47227, Korea.,Anti-Aging Research Center, Dongeui University, Busan 47340, Korea
| | - Su-Hyun Hong
- Open Laboratory for Muscular and Skeletal Disease, and Department of Biochemistry, Dongeui University College of Korean Medicine, 42 San, Yangjungdong, Busan 47227, Korea.,Anti-Aging Research Center, Dongeui University, Busan 47340, Korea
| | - Jin-Woo Jeong
- Open Laboratory for Muscular and Skeletal Disease, and Department of Biochemistry, Dongeui University College of Korean Medicine, 42 San, Yangjungdong, Busan 47227, Korea.,Anti-Aging Research Center, Dongeui University, Busan 47340, Korea
| | - Gi Young Kim
- Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National University, Jeju 63243, Korea
| | - Cheol Park
- Department of Molecular Biology, College of Natural Sciences, Dongeui University, Busan 47340, Korea
| | - Yung Hyun Choi
- Open Laboratory for Muscular and Skeletal Disease, and Department of Biochemistry, Dongeui University College of Korean Medicine, 42 San, Yangjungdong, Busan 47227, Korea.,Anti-Aging Research Center, Dongeui University, Busan 47340, Korea
| |
Collapse
|
12
|
Ahn KI, Choi EO, Kwon DH, HwangBo H, Kim MY, Kim HJ, Ji SY, Hong SH, Jeong JW, Park C, Kim ND, Kim WJ, Choi YH. Induction of apoptosis by ethanol extract of Citrus unshiu Markovich peel in human bladder cancer T24 cells through ROS-mediated inactivation of the PI3K/Akt pathway. Biosci Trends 2017; 11:565-573. [PMID: 29070760 DOI: 10.5582/bst.2017.01218] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Citrus unshiu peel has been used to prevent and treat various diseases in traditional East-Asian medicine including in Korea. Extracts of C. unshiu peel are known to have various pharmacological effects including antioxidant, anti-inflammatory, and antibacterial properties. Although the possibility of their anti-cancer activity has recently been reported, the exact mechanisms in human cancer cells have not been sufficiently studied. In this study, the inhibitory effect of ethanol extract of C. unshiu peel (EECU) on the growth of human bladder cancer T24 cells was evaluated and the underlying mechanism was investigated. The present study demonstrated that the suppression of T24 cell viability by EECU is associated with apoptosis induction. EECU-induced apoptosis was found to correlate with an activation of caspase-8, -9, and -3 in concomitance with a decrease in the expression of the inhibitor of apoptosis family of proteins and an increase in the Bax:Bcl-2 ratio accompanied by the proteolytic degradation of poly(ADP-ribose) polymerase. EECU also increased the generation of reactive oxygen species (ROS), collapse of mitochondrial membrane potential, and cytochrome c release to the cytosol, along with a truncation of Bid. In addition, EECU inactivated phosphatidylinositol 3-kinase (PI3K) as well as Akt, a downstream molecular target of PI3K, and LY294002, a specific PI3K inhibitor significantly enhanced EECU-induced apoptosis and cell viability reduction. However, N-acetyl cysteine, a general ROS scavenger, completely reversed the EECU-induced dephosphorylation of PI3K and Akt, as well as cell apoptosis. Taken together, these findings suggest that EECU inhibits T24 cell proliferation by activating intrinsic and extrinsic apoptosis pathways through a ROS-mediated inactivation of the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Kyu Im Ahn
- Open Laboratory for Muscular and Skeletal Disease, and Department of Biochemistry, Dongeui University College of Korean Medicine.,Anti-Aging Research Center, Dongeui University.,Department of Pharmacy, Molecular Inflammation Research Center for Aging Intervention, Pusan National University
| | - Eun Ok Choi
- Open Laboratory for Muscular and Skeletal Disease, and Department of Biochemistry, Dongeui University College of Korean Medicine.,Anti-Aging Research Center, Dongeui University
| | - Da He Kwon
- Open Laboratory for Muscular and Skeletal Disease, and Department of Biochemistry, Dongeui University College of Korean Medicine.,Anti-Aging Research Center, Dongeui University
| | - Hyun HwangBo
- Open Laboratory for Muscular and Skeletal Disease, and Department of Biochemistry, Dongeui University College of Korean Medicine.,Anti-Aging Research Center, Dongeui University
| | - Min Yeong Kim
- Open Laboratory for Muscular and Skeletal Disease, and Department of Biochemistry, Dongeui University College of Korean Medicine.,Anti-Aging Research Center, Dongeui University
| | - Hong Jae Kim
- Open Laboratory for Muscular and Skeletal Disease, and Department of Biochemistry, Dongeui University College of Korean Medicine.,Anti-Aging Research Center, Dongeui University
| | - Seon Yeong Ji
- Open Laboratory for Muscular and Skeletal Disease, and Department of Biochemistry, Dongeui University College of Korean Medicine.,Anti-Aging Research Center, Dongeui University
| | - Su-Hyun Hong
- Open Laboratory for Muscular and Skeletal Disease, and Department of Biochemistry, Dongeui University College of Korean Medicine.,Anti-Aging Research Center, Dongeui University
| | - Jin-Woo Jeong
- Open Laboratory for Muscular and Skeletal Disease, and Department of Biochemistry, Dongeui University College of Korean Medicine.,Anti-Aging Research Center, Dongeui University
| | - Cheol Park
- Department of Molecular Biology, College of Natural Sciences, Dongeui University
| | - Nam Deuk Kim
- Department of Pharmacy, Molecular Inflammation Research Center for Aging Intervention, Pusan National University
| | - Wun Jae Kim
- Personalized Tumor Engineering Research Center, Department of Urology, Chungbuk National University College of Medicine
| | - Yung Hyun Choi
- Open Laboratory for Muscular and Skeletal Disease, and Department of Biochemistry, Dongeui University College of Korean Medicine.,Anti-Aging Research Center, Dongeui University
| |
Collapse
|
13
|
Choi YH. Diallyl trisulfide induces apoptosis and mitotic arrest in AGS human gastric carcinoma cells through reactive oxygen species-mediated activation of AMP-activated protein kinase. Biomed Pharmacother 2017; 94:63-71. [PMID: 28753455 DOI: 10.1016/j.biopha.2017.07.055] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/01/2017] [Accepted: 07/11/2017] [Indexed: 01/08/2023] Open
Abstract
Diallyl trisulfide (DATS), one of the principal constituents of garlic oil, is a kind of organosulfur compound with high anti-cancer activity. Although inhibition of cancer cell proliferation by DATS is known to be associated with the induction of apoptosis and cell cycle arrest related to reactive oxygen species (ROS) production, it is still necessary to study the detailed mechanisms. In this study, we investigated the role of ROS on the activation of AMP-activated protein kinase (AMPK) in DATS-induced apoptosis and cell cycle arrest in AGS human gastric carcinoma cells. The results of the present study indicate that DATS inhibited proliferation of AGS cells by promoting apoptosis, and accumulating cellular portion of G2/M phase via the induction of cyclin B1 and cyclin-dependent kinase p21(WAF1/CIP1). The phosphorylation of histone H3 was also markedly increased following treatment with DATS, revealing that DATS stimulated a mitotic arrest, not the G2 phase. Furthermore, we found that DATS concurrently induced phosphorylation of AMPK; however, chemical inhibition of AMPK by compound C, an AMPK inhibitor, significantly blocked apoptosis induced by DATS, suggesting that DATS induces cytotoxicity of AGS cells through the AMPK-dependent pathway. Moreover, DATS provoked intracellular ROS generation and the loss of mitochondrial membrane potential, and in particular, when ROS production was blocked by antioxidant N-acety-l-cysteine, both AMPK activation and growth inhibition by DATS were completely abolished. Collectively, these findings suggest that DATS inhibited growth of AGS cells, which was mediated by complex interplay between cellular mechanisms governing redox homeostasis, apoptosis, and cell cycle arrest, through a ROS-dependent activation of AMPK pathway.
Collapse
Affiliation(s)
- Yung Hyun Choi
- Department of Biochemistry, Dongeui University College of Korean Medicine, Busan 47227, Republic of Korea; Anti-Aging Research Center & Blue-Bio Industry RIC, Dongeui University, Busan 47340, Republic of Korea.
| |
Collapse
|