1
|
Tao H, Wang C, Zou C, Zhu H, Zhang W. Unraveling the potential of neuroinflammation and autophagy in schizophrenia. Eur J Pharmacol 2025; 997:177469. [PMID: 40054715 DOI: 10.1016/j.ejphar.2025.177469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 02/03/2025] [Accepted: 03/04/2025] [Indexed: 03/17/2025]
Abstract
Schizophrenia (SCZ) is a complex and chronic psychiatric disorder that affects a significant proportion of the global population. Although the precise etiology of SCZ remains uncertain, recent studies have underscored the involvement of neuroinflammation and autophagy in its pathogenesis. Neuroinflammation, characterized by hyperactivated microglia and markedly elevated pro-inflammatory cytokines, has been observed in postmortem brain tissues of SCZ patients and is closely associated with disease severity. Autophagy, a cellular process responsible for eliminating damaged components and maintaining cellular homeostasis, is believed to play a pivotal role in neuronal health and the onset of SCZ. This review explores the roles and underlying mechanisms of neuroinflammation and autophagy in SCZ, with a particular focus on their intricate interplay. Additionally, we provide an overview of potential therapeutic strategies aimed at modulating neuroinflammation and autophagy, including nutritional interventions, anti-inflammatory drugs, antipsychotics, and plant-derived natural compounds. The review also addresses the dual effects of antipsychotics on autophagy. Our objective is to translate these insights into clinical practice, expanding the therapeutic options available to improve the overall health and well-being of individuals with SCZ.
Collapse
Affiliation(s)
- Hongxia Tao
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Congyin Wang
- Department of Emergency Medicine, Chengdu Fifth People's Hospital, Chengdu, Sichuan, 611130, China
| | - Chuan Zou
- Department of General Practice, Chengdu Fifth People's Hospital, Chengdu, Sichuan, 611130, China
| | - Hongru Zhu
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Wei Zhang
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
2
|
Michalak KP, Michalak AZ, Brenk-Krakowska A. Acute COVID-19 and LongCOVID syndrome - molecular implications for therapeutic strategies - review. Front Immunol 2025; 16:1582783. [PMID: 40313948 PMCID: PMC12043656 DOI: 10.3389/fimmu.2025.1582783] [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: 02/25/2025] [Accepted: 03/28/2025] [Indexed: 05/03/2025] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has been recognized not only for its acute effects but also for its ability to cause LongCOVID Syndrome (LCS), a condition characterized by persistent symptoms affecting multiple organ systems. This review examines the molecular and immunological mechanisms underlying LCS, with a particular focus on autophagy inhibition, chronic inflammation, oxidative, nitrosative and calcium stress, viral persistence and autoimmunology. Potential pathophysiological mechanisms involved in LCS include (1) autoimmune activation, (2) latent viral persistence, where SARS-CoV-2 continues to influence host metabolism, (3) reactivation of latent pathogens such as Epstein-Barr virus (EBV) or cytomegalovirus (CMV), exacerbating immune and metabolic dysregulation, and (4) possible persistent metabolic and inflammatory dysregulation, where the body fails to restore post-infection homeostasis. The manipulation of cellular pathways by SARS-CoV-2 proteins is a critical aspect of the virus' ability to evade immune clearance and establish long-term dysfunction. Viral proteins such as NSP13, ORF3a and ORF8 have been shown to disrupt autophagy, thereby impairing viral clearance and promoting immune evasion. In addition, mitochondrial dysfunction, dysregulated calcium signaling, oxidative stress, chronic HIF-1α activation and Nrf2 inhibition create a self-sustaining inflammatory feedback loop that contributes to tissue damage and persistent symptoms. Therefore understanding the molecular basis of LCS is critical for the development of effective therapeutic strategies. Targeting autophagy and Nrf2 activation, glycolysis inhibition, and restoration calcium homeostasis may provide novel strategies to mitigate the long-term consequences of SARS-CoV-2 infection. Future research should focus on personalized therapeutic interventions based on the dominant molecular perturbations in individual patients.
Collapse
Affiliation(s)
- Krzysztof Piotr Michalak
- Laboratory of Vision Science and Optometry, Physics and Astronomy Faculty, Adam Mickiewicz University in Poznań, Poznań, Poland
| | | | - Alicja Brenk-Krakowska
- Laboratory of Vision Science and Optometry, Physics and Astronomy Faculty, Adam Mickiewicz University in Poznań, Poznań, Poland
| |
Collapse
|
3
|
Michalak KP, Michalak AZ. Understanding chronic inflammation: couplings between cytokines, ROS, NO, Ca i 2+, HIF-1α, Nrf2 and autophagy. Front Immunol 2025; 16:1558263. [PMID: 40264757 PMCID: PMC12012389 DOI: 10.3389/fimmu.2025.1558263] [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: 01/10/2025] [Accepted: 03/14/2025] [Indexed: 04/24/2025] Open
Abstract
Chronic inflammation is an important component of many diseases, including autoimmune diseases, intracellular infections, dysbiosis and degenerative diseases. An important element of this state is the mainly positive feedback between inflammatory cytokines, reactive oxygen species (ROS), nitric oxide (NO), increased intracellular calcium, hypoxia-inducible factor 1-alpha (HIF-1α) stabilisation and mitochondrial oxidative stress, which, under normal conditions, enhance the response against pathogens. Autophagy and the nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated antioxidant response are mainly negatively coupled with the above-mentioned elements to maintain the defence response at a level appropriate to the severity of the infection. The current review is the first attempt to build a multidimensional model of cellular self-regulation of chronic inflammation. It describes the feedbacks involved in the inflammatory response and explains the possible pathways by which inflammation becomes chronic. The multiplicity of positive feedbacks suggests that symptomatic treatment of chronic inflammation should focus on inhibiting multiple positive feedbacks to effectively suppress all dysregulated elements including inflammation, oxidative stress, calcium stress, mito-stress and other metabolic disturbances.
Collapse
Affiliation(s)
- Krzysztof Piotr Michalak
- Laboratory of Vision Science and Optometry, Physics and Astronomy Faculty, Adam Mickiewicz University in Poznań, Poznań, Poland
| | | |
Collapse
|
4
|
Silva RCMC. The dichotomic role of cytokines in aging. Biogerontology 2024; 26:17. [PMID: 39621124 DOI: 10.1007/s10522-024-10152-4] [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: 09/04/2024] [Accepted: 10/30/2024] [Indexed: 12/11/2024]
Abstract
The chronic inflammation present in aged individuals is generally depicted as a detrimental player for longevity. Here, it is discussed several beneficial effects associated with the cytokines that are chronically elevated in inflammaging. These cytokines, such as IL-1β, type I interferons, IL-6 and TNF positively regulate macroautophagy, mitochondrial function, anti-tumor immune responses and skeletal muscle biogenesis, possibly contributing to longevity. On the other side, the detrimental and antagonistic role of these cytokines including the induction of sarcopenia, tissue damage and promotion of tumorigenesis are also discussed, underscoring the dichotomy associated with inflammaging and its players. In addition, it is discussed the role of the anti-inflammatory cytokine IL-10 and other cytokines that affect aging in a more linear way, such as IL-11, which promotes senescence, and IL-4 and IL-15, which promotes longevity. It is also discussed more specific regulators of aging that are downstream cytokines-mediated signaling.
Collapse
|
5
|
Zhang G, Wang Q, Yuan R, Zhang Y, Chen K, Yu J, Ye T, Jia X, Zhou Y, Li G, Chen K. Oncolytic vaccinia virus harboring aphrocallistes vastus lectin exerts anti-tumor effects by directly oncolysis and inducing immune response through enhancing ROS in human ovarian cancer. Biochem Biophys Res Commun 2024; 730:150355. [PMID: 38996784 DOI: 10.1016/j.bbrc.2024.150355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 07/04/2024] [Indexed: 07/14/2024]
Abstract
Aphrocallistes vastus lectin (AVL) is a Ca2+ dependent C-type lectin produced by sponges. Previous studies have demonstrated that oncolytic vaccinia virus harboring AVL (oncoVV-AVL) effectively triggers cell death in various tumors. However, the effects of oncoVV-AVL on human ovarian cancer (OV) remain unknown. This study aims to investigate the mechanism-of-action of oncoVV-AVL in human OV cell lines and in tumor-bearing nude mice. We found that oncoVV-AVL could directly induce apoptosis and autophagy in ovarian cancer cells. Additionally, our results showed that oncoVV-AVL increased the serum levels of mouse IFN-γ (mIFN-γ), leading to the activation of M1-polarized macrophages. Conversely, NADPH, a reducing agent by providing reducing equivalents, reduced the production of mIFN-γ, and suppressed M1-polarization of macrophage. Based on these findings, we propose that oncoVV-AVL not only contributes to direct cytolysis, but also enhances host immune response by promoting ROS levels.
Collapse
Affiliation(s)
- Guohui Zhang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Qiang Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Rentao Yuan
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yanan Zhang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Ke Chen
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Jianlei Yu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Ting Ye
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiaoyuan Jia
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yanrong Zhou
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Gongchu Li
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China; Hangzhou Gongchu Biotechnology Co., Ltd., Hangzhou, China.
| | - Kan Chen
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China.
| |
Collapse
|
6
|
Zhu X, Wang L, Wang K, Yao Y, Zhou F. Erdafitinib promotes ferroptosis in human uveal melanoma by inducing ferritinophagy and lysosome biogenesis via modulating the FGFR1/mTORC1/TFEB signaling axis. Free Radic Biol Med 2024; 222:552-568. [PMID: 38971541 DOI: 10.1016/j.freeradbiomed.2024.07.002] [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: 05/14/2024] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Uveal melanoma (UM) is a rare yet lethal primary intraocular malignancy affecting adults. Analysis of data from The Cancer Genome Atlas (TCGA) database revealed that FGFR1 expression was increased in UM tumor tissues and was linked to aggressive behavior and a poor prognosis. This study assessed the anti-tumor effects of Erdafitinib, a selective pan-FGFR inhibitor, in both in vitro and in vivo UM models. Erdafitinib exhibited a robust anti-cancer activity in UM through inducing ferroptosis in the FGFR1-dependent manner. Transcriptomic data revealed that Erdafitinib mediated its anti-cancer effects via modulating the ferritinophagy/lysosome biogenesis. Subsequent research revealed that Erdafitinib exerted its effects by reducing the expression of FGFR1 and inhibiting the activity of mTORC1 in UM cells. Concurrently, it enhanced the dephosphorylation, nuclear translocation, and transcriptional activity of TFEB. The aggregation of TFEB in nucleus triggered FTH1-dependent ferritinophagy, leading to lysosomal activation and iron overload. Conversely, the overexpression of FGFR1 served to mitigate the effects of Erdafitinib on ferritinophagy, lysosome biogenesis, and the activation of the mTORC1/TFEB signaling pathway. In vivo experiments have convincingly shown that Erdafitinib markedly curtails tumor growth in an UM xenograft mouse model, an effect that is closely correlated with a decrease in FGFR1 expression levels. The present study is the first to demonstrate that Erdafitinib powerfully induces ferroptosis in UM by orchestrating the ferritinophagy and lysosome biogenesis via modulating the FGFR1/mTORC1/TFEB signaling. Consequently, Erdafitinib emerges as a strong candidate for clinical trial investigation, and FGFR1 emerges as a novel and promising therapeutic target in the treatment of UM.
Collapse
Affiliation(s)
- Xue Zhu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, Jiangsu Province, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, China
| | - Ling Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, Jiangsu Province, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, China
| | - Ke Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, Jiangsu Province, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, China.
| | - Ying Yao
- Department of Pharmacy, Wuxi Maternity and Child Health Care Hospital, Women's Hospital of Jiangnan University, Jiangnan University, Wuxi, 214002, Jiangsu Province, China.
| | - Fanfan Zhou
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
7
|
Walweel N, Aydin O. Enhancing Therapeutic Efficacy in Cancer Treatment: Integrating Nanomedicine with Autophagy Inhibition Strategies. ACS OMEGA 2024; 9:27832-27852. [PMID: 38973850 PMCID: PMC11223161 DOI: 10.1021/acsomega.4c02234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/01/2024] [Accepted: 05/30/2024] [Indexed: 07/09/2024]
Abstract
The complicated stepwise lysosomal degradation process known as autophagy is in charge of destroying and eliminating damaged organelles and defective cytoplasmic components. This mechanism promotes metabolic adaptability and nutrition recycling. Autophagy functions as a quality control mechanism in cells that support homeostasis and redox balance under normal circumstances. However, the role of autophagy in cancer is controversial because, mostly depending on the stage of the tumor, it may either suppress or support the disease. While autophagy delays the onset of tumors and slows the dissemination of cancer in the early stages of tumorigenesis, numerous studies demonstrate that autophagy promotes the development and spread of tumors as well as the evolution and development of resistance to several anticancer drugs in advanced cancer stages. In this Review, we primarily emphasize the therapeutic role of autophagy inhibition in improving the treatment of multiple cancers and give a broad overview of how its inhibition modulates cancer responses. There have been various attempts to inhibit autophagy, including the use of autophagy inhibitor drugs, gene silencing therapy (RNA interference), and nanoparticles. In this Review, all these topics are thoroughly covered and illustrated by recent studies and field investigations.
Collapse
Affiliation(s)
- Nada Walweel
- Department
of Biomedical Engineering, Erciyes University, Kayseri 38039, Turkey
- NanoThera
Lab, ERFARMA-Drug Application and Research Center, Erciyes University, Kayseri 38280, Turkey
| | - Omer Aydin
- Department
of Biomedical Engineering, Erciyes University, Kayseri 38039, Turkey
- NanoThera
Lab, ERFARMA-Drug Application and Research Center, Erciyes University, Kayseri 38280, Turkey
- ERNAM-Nanotechnology
Research and Application Center, Erciyes
University, Kayseri 38039, Turkey
- ERKAM-Clinical-Engineering
Research and Implementation Center, Erciyes
University, Kayseri 38030, Turkey
| |
Collapse
|
8
|
Abd El-Aziz YS, Toit-Thompson TD, McKay MJ, Molloy MP, Stoner S, McDowell B, Moon E, Sioson L, Sheen A, Chou A, Gill AJ, Jansson PJ, Sahni S. Novel combinatorial autophagy inhibition therapy for triple negative breast cancers. Eur J Pharmacol 2024; 973:176568. [PMID: 38604544 DOI: 10.1016/j.ejphar.2024.176568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND Triple negative breast cancer (TNBC) has the worst prognosis among breast cancer subtypes. It is characterized by lack of estrogen, progesterone and human epidermal growth factor 2 receptors, and thus, have limited therapeutic options. Autophagy has been found to be correlated with poor prognosis and aggressive behaviour in TNBC. This study aimed to target autophagy in TNBC via a novel approach to inhibit TNBC progression. METHODS Immunoblotting and confocal microscopy were carried out to examine the effect of tumor microenvironmental stressors on autophagy. Cellular proliferation and migration assays were used to test the effect of different autophagy inhibitors and standard chemotherapy alone or in combination. In vivo xenograft mouse model was utilized to assess the effect of autophagy inhibitors alone or in combination with Paclitaxel. High resolution mass spectrometry based proteomic analysis was performed to explore the mechanisms behind chemoresistance in TNBC. Lastly, immunohistochemistry was done to assess the correlation between autophagy related proteins and clinical characteristics in TNBC tissue specimens. RESULTS Metabolic stressors were found to induce autophagy in TNBC cell lines. Autophagy initiation inhibitors, SAR405 and MRT68921, showed marked synergy in their anti-proliferative activity in both chemosensitive and chemoresistant TNBC cell models. Paradoxically, positive expression of autophagosome marker LC3 was shown to be associated with better overall survival of TNBC patients. CONCLUSION In this study, a novel combination between different autophagy inhibitors was identified which inhibited tumor cell proliferation in both chemosensitive and chemoresistant TNBC cells and could result in development of a novel treatment modality against TNBC.
Collapse
Affiliation(s)
- Yomna S Abd El-Aziz
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Australia; Kolling Institute of Medical Research, University of Sydney, Australia; Oral Pathology Department, Faculty of Dentistry, Tanta University, Tanta, Egypt
| | - Taymin du Toit-Thompson
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Australia; Kolling Institute of Medical Research, University of Sydney, Australia
| | - Matthew J McKay
- Kolling Institute of Medical Research, University of Sydney, Australia
| | - Mark P Molloy
- Kolling Institute of Medical Research, University of Sydney, Australia
| | - Shihani Stoner
- Kolling Institute of Medical Research, University of Sydney, Australia
| | - Betty McDowell
- NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, Sydney, Australia
| | - Elizabeth Moon
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Australia; Kolling Institute of Medical Research, University of Sydney, Australia
| | - Loretta Sioson
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Australia; Kolling Institute of Medical Research, University of Sydney, Australia; NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, Sydney, Australia
| | - Amy Sheen
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Australia; Kolling Institute of Medical Research, University of Sydney, Australia; NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, Sydney, Australia
| | - Angela Chou
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Australia; Kolling Institute of Medical Research, University of Sydney, Australia; NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, Sydney, Australia
| | - Anthony J Gill
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Australia; Kolling Institute of Medical Research, University of Sydney, Australia; NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, Sydney, Australia
| | - Patric J Jansson
- Kolling Institute of Medical Research, University of Sydney, Australia; Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Sumit Sahni
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Australia; Kolling Institute of Medical Research, University of Sydney, Australia.
| |
Collapse
|
9
|
Lee YS, Mun JG, Park SY, Hong DY, Kim HY, Kim SJ, Lee SB, Jang JH, Han YH, Kee JY. Saikosaponin D Inhibits Lung Metastasis of Colorectal Cancer Cells by Inducing Autophagy and Apoptosis. Nutrients 2024; 16:1844. [PMID: 38931199 PMCID: PMC11206761 DOI: 10.3390/nu16121844] [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: 05/17/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Saikosaponin D (SSD), derived from Bupleurum falcatum L., has various pharmacological properties, including immunoregulatory, anti-inflammatory, and anti-allergic effects. Several studies have investigated the anti-tumor effects of SSD on cancer in multiple organs. However, its role in colorectal cancer (CRC) remains unclear. Therefore, this study aimed to elucidate the suppressive effects of SSD on CRC cell survival and metastasis. SSD reduced the survival and colony formation ability of CRC cells. SSD-induced autophagy and apoptosis in CRC cells were measured using flow cytometry. SSD treatment increased LC3B and p62 autophagic factor levels in CRC cells. Moreover, SSD-induced apoptosis occurred through the cleavage of caspase-9, caspase-3, and PARP, along with the downregulation of the Bcl-2 family. In the in vivo experiment, a reduction in the number of metastatic tumor nodules in the lungs was observed after the oral administration of SSD. Based on these results, SSD inhibits the metastasis of CRC cells to the lungs by inducing autophagy and apoptosis. In conclusion, SSD suppressed the proliferation and metastasis of CRC cells, suggesting its potential as a novel substance for the metastatic CRC treatment.
Collapse
Affiliation(s)
- Yoon-Seung Lee
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Jeonbuk, Republic of Korea
| | - Jeong-Geon Mun
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Jeonbuk, Republic of Korea
| | - Shin-Young Park
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Jeonbuk, Republic of Korea
| | - Dah Yun Hong
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Jeonbuk, Republic of Korea
| | - Ho-Yoon Kim
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Jeonbuk, Republic of Korea
| | - Su-Jin Kim
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Jeonbuk, Republic of Korea
| | - Sun-Bin Lee
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Jeonbuk, Republic of Korea
| | - Jeong-Ho Jang
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Jeonbuk, Republic of Korea
| | - Yo-Han Han
- Department of Microbiology, Wonkwang University School of Medicine, Iksan 54538, Jeonbuk, Republic of Korea
| | - Ji-Ye Kee
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Jeonbuk, Republic of Korea
| |
Collapse
|
10
|
Kim NY, Shivanne Gowda SG, Lee SG, Sethi G, Ahn KS. Cannabidiol induces ERK activation and ROS production to promote autophagy and ferroptosis in glioblastoma cells. Chem Biol Interact 2024; 394:110995. [PMID: 38583854 DOI: 10.1016/j.cbi.2024.110995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/27/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
Small molecule-driven ERK activation is known to induce autophagy and ferroptosis in cancer cells. Herein the effect of cannabidiol (CBD), a phytochemical derived from Cannabis sativa, on ERK-driven autophagy and ferroptosis has been demonstrated in glioblastoma (GBM) cells (U87 and U373 cells). CBD imparted significant cytotoxicity in GBM cells, induced activation of ERK (not JNK and p38), and increased intracellular reactive oxygen species (ROS) levels. It increased the autophagy-related proteins such as LC3 II, Atg7, and Beclin-1 and modulated the expression of ferroptosis-related proteins such as glutathione peroxidase 4 (GPX4), SLC7A11, and TFRC. CBD significantly elevated the endoplasmic reticulum stress, ROS, and iron load, and decreased GSH levels. Inhibitors of autophagy (3-MA) and ferroptosis (Fer-1) had a marginal effect on CBD-induced autophagy/ferroptosis. Treatment with N-acetyl-cysteine (antioxidant) or PD98059 (ERK inhibitor) partly reverted the CBD-induced autophagy/ferroptosis by decreasing the activation of ERK and the production of ROS. Overall, CBD induced autophagy and ferroptosis through the activation of ERK and generation of ROS in GBM cells.
Collapse
Affiliation(s)
- Na Young Kim
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| | | | - Seok-Geun Lee
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| |
Collapse
|
11
|
Asano S, Ono A, Baba K, Uehara T, Sakamoto K, Hayata-Takano A, Nakazawa T, Yanamoto S, Tanimoto K, Hashimoto H, Ago Y. Blockade of vasoactive intestinal peptide receptor 2 (VIPR2) signaling suppresses cyclin D1-dependent cell-cycle progression in MCF-7 cells. J Pharmacol Sci 2024; 154:139-147. [PMID: 38395514 DOI: 10.1016/j.jphs.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/28/2023] [Accepted: 01/05/2024] [Indexed: 02/25/2024] Open
Abstract
Vasoactive intestinal peptide (VIP) receptor 2 (VIPR2) is a G protein-coupled receptor that binds to Gαs, Gαi, and Gαq proteins to regulate various downstream signaling molecules, such as protein kinase A (PKA), phosphatidylinositol 3-kinase (PI3K), and phospholipase C. In this study, we examined the role of VIPR2 in cell cycle progression. KS-133, a newly developed VIPR2-selective antagonist peptide, attenuated VIP-induced cell proliferation in MCF-7 cells. The percentage of cells in the S-M phase was decreased in MCF-7 cells treated with KS-133. KS-133 in the presence of VIP decreased the phosphorylation of extracellular signal-regulated kinase (ERK), AKT, and glycogen synthase kinase-3β (GSK3β), resulting in a decrease in cyclin D1 levels. In MCF-7 cells stably-expressing VIPR2, KS-133 decreased PI3K activity and cAMP levels. Treatment with the ERK-specific kinase (MEK) inhibitor U0126 and the class I PI3K inhibitor ZSTK474 decreased the percentage of cells in the S phase. KS-133 reduced the percentage of cells in the S phase more than treatment with U0126 or ZSTK474 alone and did not affect the effect of the mixture of these inhibitors. Our findings suggest that VIPR2 signaling regulates cyclin D1 levels through the cAMP/PKA/ERK and PI3K/AKT/GSK3β pathways, and mediates the G1/S transition to control cell proliferation.
Collapse
Affiliation(s)
- Satoshi Asano
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan; School of Dentistry, Hiroshima University, Hiroshima, 734-8553, Japan.
| | - Ami Ono
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan; Department of Orthodontics and Craniofacial Developmental Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Kaede Baba
- School of Dentistry, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Teru Uehara
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan; Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Kotaro Sakamoto
- Research & Development Department, Ichimaru Pharcos Company Limited, 318-1 Asagi, Motosu, Gifu, 501-0475, Japan
| | - Atsuko Hayata-Takano
- Department of Pharmacology, Graduate School of Dentistry, Osaka University, Suita, Osaka, 565-0871, Japan; Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University, University of Fukui, Osaka, 565-0871, Japan
| | - Takanobu Nakazawa
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Laboratory of Molecular Biology, Department of Bioscience, Graduate School of Life Sciences, Tokyo University of Agriculture, Tokyo, 156-8502, Japan
| | - Souichi Yanamoto
- School of Dentistry, Hiroshima University, Hiroshima, 734-8553, Japan; Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Kotaro Tanimoto
- School of Dentistry, Hiroshima University, Hiroshima, 734-8553, Japan; Department of Orthodontics and Craniofacial Developmental Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University, University of Fukui, Osaka, 565-0871, Japan; Division of Bioscience, Institute for Datability Science, Osaka University, Osaka, 565-0871, Japan; Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, 565-0871, Japan; Department of Molecular Pharmaceutical Science, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Yukio Ago
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan; School of Dentistry, Hiroshima University, Hiroshima, 734-8553, Japan.
| |
Collapse
|
12
|
Li Q, Lin Y, Liang G, Xiao N, Zhang H, Yang X, Yang J, Liu A. Autophagy and Senescence: The Molecular Mechanisms and Implications in Liver Diseases. Int J Mol Sci 2023; 24:16880. [PMID: 38069199 PMCID: PMC10706096 DOI: 10.3390/ijms242316880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 11/21/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
The liver is the primary organ accountable for complex physiological functions, including lipid metabolism, toxic chemical degradation, bile acid synthesis, and glucose metabolism. Liver function homeostasis is essential for the stability of bodily functions and is involved in the complex regulation of the balance between cell proliferation and cell death. Cell proliferation-halting mechanisms, including autophagy and senescence, are implicated in the development of several liver diseases, such as cholestasis, viral hepatitis, nonalcoholic fatty liver disease, liver fibrosis, and hepatocellular carcinoma. Among various cell death mechanisms, autophagy is a highly conserved and self-degradative cellular process that recycles damaged organelles, cellular debris, and proteins. This process also provides the substrate for further metabolism. A defect in the autophagy machinery can lead to premature diseases, accelerated aging, inflammatory state, tumorigenesis, and cellular senescence. Senescence, another cell death type, is an active player in eliminating premalignant cells. At the same time, senescent cells can affect the function of neighboring cells by secreting the senescence-associated secretory phenotype and induce paracrine senescence. Autophagy can promote and delay cellular senescence under different contexts. This review decodes the roles of autophagy and senescence in multiple liver diseases to achieve a better understanding of the regulatory mechanisms and implications of autophagy and senescence in various liver diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Anding Liu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan 430100, China; (Q.L.); (Y.L.); (G.L.); (N.X.); (H.Z.); (X.Y.); (J.Y.)
| |
Collapse
|
13
|
Slimani W, Maioli M, Cruciani S, Zerizer S, Santaniello S, Kabouche Z, Coradduzza D, Chessa M, Fancello S, Migheli R, Serra PA, D'hallewin G. Antioxidant, Anti-Inflammatory and Anti-Proliferative Properties of Stachys circinata on HepG2 and MCF7 Cells. PLANTS (BASEL, SWITZERLAND) 2023; 12:2272. [PMID: 37375897 DOI: 10.3390/plants12122272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023]
Abstract
According to the WHO, the overall age-standardized cancer rate keeps declining, and the number of cases diagnosed each year increases, remaining among the leading causes of death in 91 out of 172 recorded countries. In this context, novel cancer prediction and therapeutic protocols are compulsory. The effect of a Stachys circinata L'Hér dichloromethane extract (ScDME) on cell redox homeostasis and tumor proliferation was investigated. HepG2 cell feedback mechanisms to oxidative stress exposure were evaluated by determining catalase (CAT) and reduced glutathione (GSH), following the supply with ScDME (0.0-5.7 µg/µL). Cytotoxicity of ScDME against the human umbilical vein endothelial cell (HUVEC) and two human cancer cell lines (breast: MCF7; liver: HepG2) was evaluated by the MTT assay. H2O2-stressed HepG2 cells supplied with the S. circinata extracts exhibited significantly increased CAT and GSH activity as compared to unsupplied ones. The anti-inflammatory activity of the extracts was evaluated by real time-qPCR on IL-1, IL-6 and TNF-α expression. As a result, this research points out that S. circinata dichloromethane extract owns anti-inflammatory and anti-proliferative properties against MCF7 and HepG2 cells and activates CAT and GSH of the HepG2 cells' antioxidant enzyme system.
Collapse
Affiliation(s)
- Wassila Slimani
- Laboratoire d'Obtention de Substances Thérapeutiques (L.O.S.T), Département de Chimie, Université des Frères Mentouri-Constantine, Constantine 25000, Algeria
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
| | - Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
| | - Sakina Zerizer
- Laboratoire d'Obtention de Substances Thérapeutiques (L.O.S.T), Département de Chimie, Université des Frères Mentouri-Constantine, Constantine 25000, Algeria
| | - Sara Santaniello
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
| | - Zahia Kabouche
- Laboratoire d'Obtention de Substances Thérapeutiques (L.O.S.T), Département de Chimie, Université des Frères Mentouri-Constantine, Constantine 25000, Algeria
| | - Donatella Coradduzza
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
| | - Mario Chessa
- Department of Medicine, Surgery and Pharmacy, Sassari University, Viale San Pietro 43/b, 07100 Sassari, Italy
| | - Silvia Fancello
- Department of Medicine, Surgery and Pharmacy, Sassari University, Viale San Pietro 43/b, 07100 Sassari, Italy
| | - Rossana Migheli
- Department of Medicine, Surgery and Pharmacy, Sassari University, Viale San Pietro 43/b, 07100 Sassari, Italy
| | - Pier Andrea Serra
- Department of Medicine, Surgery and Pharmacy, Sassari University, Viale San Pietro 43/b, 07100 Sassari, Italy
| | - Guy D'hallewin
- Institute of Sciences of Food Production, National Research Council, Traversa la Crucca, 3. Loc Baldinca Li Punti, 07100 Sassari, Italy
| |
Collapse
|
14
|
Barnard SJ, Haunschild J, Heiser L, Dieterlen MT, Klaeske K, Borger MA, Etz CD. Apoptotic Cell Death in Bicuspid-Aortic-Valve-Associated Aortopathy. Int J Mol Sci 2023; 24:ijms24087429. [PMID: 37108591 PMCID: PMC10138609 DOI: 10.3390/ijms24087429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/12/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
The bicuspid aortic valve (BAV) is the most common cardiovascular congenital abnormality and is frequently associated with proximal aortopathy. We analyzed the tissues of patients with bicuspid and tricuspid aortic valve (TAV) regarding the protein expression of the receptor for advanced glycation products (RAGE) and its ligands, the advanced glycation end products (AGE), as well as the S100 calcium-binding protein A6 (S100A6). Since S100A6 overexpression attenuates cardiomyocyte apoptosis, we investigated the diverse pathways of apoptosis and autophagic cell death in the human ascending aortic specimen of 57 and 49 patients with BAV and TAV morphology, respectively, to identify differences and explanations for the higher risk of patients with BAV for severe cardiovascular diseases. We found significantly increased levels of RAGE, AGE and S100A6 in the aortic tissue of bicuspid patients which may promote apoptosis via the upregulation of caspase-3 activity. Although increased caspase-3 activity was not detected in BAV patients, increased protein expression of the 48 kDa fragment of vimentin was detected. mTOR as a downstream protein of Akt was significantly higher in patients with BAV, whereas Bcl-2 was increased in patients with TAV, assuming a better protection against apoptosis. The autophagy-related proteins p62 and ERK1/2 were increased in patients with BAV, assuming that cells in bicuspid tissue are more likely to undergo apoptotic cell death leading to changes in the wall and finally to aortopathies. We provide first-hand evidence of increased apoptotic cell death in the aortic tissue of BAV patients which may thus provide an explanation for the increased risk of structural aortic wall deficiency possibly underlying aortic aneurysm formation or acute dissection.
Collapse
Affiliation(s)
- Sarah J Barnard
- Heisenberg Working Group, Saxonian Incubator for Clinical Translation, Philipp-Rosenthal-Str. 55, 04103 Leipzig, Germany
| | - Josephina Haunschild
- Heisenberg Working Group, Saxonian Incubator for Clinical Translation, Philipp-Rosenthal-Str. 55, 04103 Leipzig, Germany
- University Department for Cardiac Surgery, Heart Center Leipzig, 04289 Leipzig, Germany
| | - Linda Heiser
- University Department for Cardiac Surgery, Heart Center Leipzig, 04289 Leipzig, Germany
| | - Maja T Dieterlen
- University Department for Cardiac Surgery, Heart Center Leipzig, 04289 Leipzig, Germany
| | - Kristin Klaeske
- University Department for Cardiac Surgery, Heart Center Leipzig, 04289 Leipzig, Germany
| | - Michael A Borger
- University Department for Cardiac Surgery, Heart Center Leipzig, 04289 Leipzig, Germany
| | - Christian D Etz
- Heisenberg Working Group, Saxonian Incubator for Clinical Translation, Philipp-Rosenthal-Str. 55, 04103 Leipzig, Germany
- University Department for Cardiac Surgery, Heart Center Leipzig, 04289 Leipzig, Germany
| |
Collapse
|
15
|
Mohamed OS, Abdel Baky NA, Sayed-Ahmed MM, Al-Najjar AH. Lactoferrin alleviates cyclophosphamide induced-nephropathy through suppressing the orchestration between Wnt4/β-catenin and ERK1/2/NF-κB signaling and modulating klotho and Nrf2/HO-1 pathway. Life Sci 2023; 319:121528. [PMID: 36828132 DOI: 10.1016/j.lfs.2023.121528] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023]
Abstract
AIMS Cyclophosphamide is an alkylating agent with vast arrays of therapeutic activity. Currently, its medical use is limited due to its numerous adverse events, including nephrotoxicity. This study aimed to follow the molecular mechanisms behind the potential renoprotective action of lactoferrin (LF) against cyclophosphamide (CP)-induced renal injury. MATERIALS AND METHODS For fulfillment of our aim, Spragw-Dwaly rats were orally administrated LF (300 mg/kg) for seven consecutive days, followed by a single intraperitoneal injection of CP (150 mg/kg). KEY FINDINGS Treatment of CP-injured rats with LF significantly reduced the elevated creatinine and blood urea nitrogen (BUN), markedly upregulated Nrf2/HO-1 signaling with consequent increase in renal total antioxidant capacity (TAC) and decrease in renal malondialdehyde (MDA) level. Furthermore, LF treatment significantly reduced the elevated renal p-ERK1/2 expression, tumor necrosis factor-α (TNFα), interleukin-6 (IL-6), nuclear factor-kappa B (NF-κB) levels in CP-treated animals. Interestingly, LF treatment downregulated Wnt4/β-catenin signaling and increased both renal klotho gene expression and serum klotho level. Furthermore, LF treatment reduced apoptosis in kidney tissue via suppressing GSK-3β expression and modulating caspase-3 and Bcl2 levels. Histopathological examination of kidney tissue confirmed the protective effect of LF against CP-induced renal injury. SIGNIFICANCE The present findings document the renoprotective effect of LF against CP-induced nephropathy, which may be mediated via suppressing ERK1/2/ NF-κB and Wnt4/β-catenin trajectories and enhancing klotho expression and Nrf2/HO-1 signaling.
Collapse
Affiliation(s)
- Ola S Mohamed
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Nayira A Abdel Baky
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt.
| | - Mohamed M Sayed-Ahmed
- Pharmacology and Experimental Oncology Unit, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Aya H Al-Najjar
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
16
|
du Plessis M, Davis TA, Olivier DW, de Villiers WJS, Engelbrecht AM. A functional role for Serum Amyloid A in the molecular regulation of autophagy in breast cancer. Front Oncol 2022; 12:1000925. [PMID: 36248994 PMCID: PMC9562844 DOI: 10.3389/fonc.2022.1000925] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/07/2022] [Indexed: 11/13/2022] Open
Abstract
It has been established that the acute phase protein, Serum amyloid A (SAA), which is usually synthesized by the liver, is also synthesized by cancer cells and cancer-associated cells in the tumor microenvironment. SAA also activates modulators of autophagy, such as the PI3K/Akt and MAPK signaling pathways. However, the role of SAA in autophagy in breast cancer still remains to be elucidated. The aim of this study was to investigate the role of SAA in the regulation of signaling pathways and autophagy in in vitro and in vivo models of breast cancer. The MDA-MB-231 and MCF7 cell lines were transiently transfected to overexpress SAA1. A tumor-bearing SAA1/2 knockout mouse model was also utilized in this study. SAA1 overexpression activated ERK signaling in the MDA-MB-231 cells, downregulated the PI3K pathway protein, PKB/Akt, in the MCF7 cell line, while SAA1/2 knockout also inhibited Akt. Furthermore, SAA1 overexpression in vitro downregulated autophagy, while the expression of SQSTM1/p62 was increased in the MCF7 cells, and SAA1/2 knockout induced autophagy in vivo. SAA overexpression in the MDA-MB-231 and MCF7 cells resulted in an increase in cell viability and increased the expression of the proliferation marker, MCM2, in the MCF7 cells. Furthermore, knockout of SAA1/2 resulted in an altered inflammatory profile, evident in the decrease of plasma IL-1β, IL-6 and IL-10, while increasing the plasma levels of MCP-1 and TNF-α. Lastly, SAA1/2 knockout promoted resistance to apoptosis and necrosis through the regulation of autophagy. SAA thus regulates autophagy in breast cancer cells to promote tumorigenesis.
Collapse
Affiliation(s)
- Manisha du Plessis
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
- *Correspondence: Manisha du Plessis,
| | - Tanja Andrea Davis
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Daniel Wilhelm Olivier
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Willem Johan Simon de Villiers
- Department of Internal Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
- African Cancer Institute (ACI), Department of Global Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
17
|
Luo ML, Huang W, Zhu HP, Peng C, Zhao Q, Han B. Advances in indole-containing alkaloids as potential anticancer agents by regulating autophagy. Biomed Pharmacother 2022; 149:112827. [PMID: 35316753 DOI: 10.1016/j.biopha.2022.112827] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/03/2022] [Accepted: 03/14/2022] [Indexed: 11/02/2022] Open
Abstract
Cancer is a leading cause of death worldwide, and cancer development is often associated with disturbances in the autophagy process. Autophagy is a catabolic process involved in many physiological processes, crucial for cell growth and survival. It is an intracellular lysosomal/vacuolar degradation system. In this system, inner cytoplasmic cell membrane is degraded by lysosomal hydrolases, and the products are released back into the cytoplasm. Indole alkaloids are natural products extensively found in nature and have been proven to possess various pharmacological activities. In recent years, pharmacological studies have demonstrated another potential of indole alkaloids, autophagy regulation. The regulation may contribute to the efficacy of indole alkaloids in preventing and treating cancer. This review summarizes the current understanding of indole alkaloids' effect on tumor cells and autophagy. Then, we focus on mechanisms by which indole alkaloids can target the autophagy process associated with cancer, including the PI3K/Akt/mTOR signaling pathway, MAPK signaling pathway, ROS signaling pathway, Beclin-1, and so on. Literature has been surveyed primarily from 2009 to Nov. 2021, and some semisynthetic or fully synthetic indole derivatives are also discussed.
Collapse
Affiliation(s)
- Meng-Lan Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong-Ping Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
18
|
Zeng M, Li Q, Chen J, Huang W, Liu J, Wang C, Huang M, Li H, Zhou S, Xie M, Zeng K. The Fgl2 interaction with Tyrobp promotes the proliferation of cutaneous squamous cell carcinoma by regulating ERK-dependent autophagy. Int J Med Sci 2022; 19:195-204. [PMID: 34975313 PMCID: PMC8692121 DOI: 10.7150/ijms.66929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/30/2021] [Indexed: 11/05/2022] Open
Abstract
Human fibroleukin 2 (Fgl2), a member of the fibrinogen superfamily, can cleave prothrombin to generate thrombin or is secreted in a soluble form as a new type of effector of Tregs with immunomodulatory functions. However, there is little research on the role of Fgl2 in cutaneous squamous cell carcinoma (CSCC) growth. We examined the expression of Fgl2 in samples from CSCC patients and CSCC cell lines. Then, the effect of Fgl2 on CSCC was evaluated in vitro and in animals. Regulation of autophagy by Fgl2 was explored in CSCC. Coimmunoprecipitation (Co-IP) and immunofluorescence colocalization experiments were conducted to identify the regulatory effect of Fgl2 on the downstream protein Tyrobp. Then, gain- or loss-of-function analyses and evaluation of Tyrobp expression were performed to validate its role in autophagy and proliferation promoted by Fgl2. Here, our study demonstrated that Fgl2 promoted the proliferation of CSCC cells in vitro and in vivo. Knocking down Fgl2 reduced CSCC cell proliferation and inhibited autophagy in CSCC. Mechanistically, Fgl2 interacted with Tyrobp and promoted ERK-dependent autophagy, resulting in the proliferation of CSCC cells. Our study suggested that Fgl2 could be a promising prognostic biomarker and useful therapeutic target for CSCC.
Collapse
Affiliation(s)
- Mei Zeng
- Department of Dermatology, Huizhou Municipal Central Hospital, Huizhou 516000, Guangdong, People's Republic of China.,Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, People's Republic of China
| | - Qingxiang Li
- Department of Dermatology, Huizhou Municipal Central Hospital, Huizhou 516000, Guangdong, People's Republic of China
| | - Junzhao Chen
- Department of Dermatology, Huizhou Municipal Central Hospital, Huizhou 516000, Guangdong, People's Republic of China
| | - Wenfu Huang
- Department of Dermatology, Huizhou Municipal Central Hospital, Huizhou 516000, Guangdong, People's Republic of China
| | - Jinhua Liu
- Department of Dermatology, Huizhou Municipal Central Hospital, Huizhou 516000, Guangdong, People's Republic of China
| | - Cuiyan Wang
- Department of Dermatology, Huizhou Municipal Central Hospital, Huizhou 516000, Guangdong, People's Republic of China
| | - Manni Huang
- Department of Dermatology, Huizhou Municipal Central Hospital, Huizhou 516000, Guangdong, People's Republic of China
| | - Hui Li
- Department of Dermatology, Huizhou Municipal Central Hospital, Huizhou 516000, Guangdong, People's Republic of China
| | - Shu Zhou
- Department of Dermatology, Huizhou Municipal Central Hospital, Huizhou 516000, Guangdong, People's Republic of China
| | - Miaoying Xie
- Department of Dermatology, Huizhou Municipal Central Hospital, Huizhou 516000, Guangdong, People's Republic of China
| | - Kang Zeng
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, People's Republic of China
| |
Collapse
|
19
|
Li M, Zhao X, Yong H, Shang B, Lou W, Wang Y, Bai J. FBXO22 Promotes Growth and Metastasis and Inhibits Autophagy in Epithelial Ovarian Cancers via the MAPK/ERK Pathway. Front Pharmacol 2021; 12:778698. [PMID: 34950036 PMCID: PMC8688818 DOI: 10.3389/fphar.2021.778698] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/15/2021] [Indexed: 12/13/2022] Open
Abstract
E3 ubiquitin ligase F-box only protein 22 (FBXO22), which targets the key regulators of cellular activities for ubiquitylation and degradation, plays an important role in tumorigenesis and metastasis. However, the function of FBXO22 in epithelial ovarian cancers has not been reported. This study aims to explore the biological function of FBXO22 in epithelial ovarian cancers progression and metastasis and its specific regulation mechanism. Immunohistochemistry analysis of tissue microarray was performed to evaluate the expression of FBXO22 in epithelial ovarian cancers patients. The proliferative ability of epithelial ovarian cancers cells was examined by the CCK8. The metastasis ability was detected by the wound healing assay, migration and invasion assays. Western blot was used to verify the relationship between FBXO22 expression and mitogen-activated protein kinase related proteins. Autophagic flux was detected by electron microscopy, mRFP-GFP-LC3 adenovirus, lysosomal tracker and western blot. For in vivo experiments, the effect of FBXO22 on epithelial ovarian cancers resistance was observed in a xenograft tumor model and a metastatic mice model. We found that FBXO22 expression was significantly increased in epithelial ovarian cancers tissues and was closely correlated with clinical pathological factors. As a result, we found that FBXO22 promoted the growth and metastasis, as well as inhibited the autophagy flux. In addition, we identified that FBXO22 performed these functions via the MAPK/ERK pathway. Our results first reported the function of FBXO22 in epithelial ovarian cancer and the correlation between FBXO22 and autophagy, suggesting FBXO22 as a novel target of epithelial ovarian cancers assessment and treatment.
Collapse
Affiliation(s)
- Minle Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Xue Zhao
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Hongmei Yong
- Department of Oncology, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, China
| | - Bingqing Shang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Weihua Lou
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - You Wang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
20
|
Ebrahim HA, El-Gamal R, Sherif RN. Intermittent Fasting Attenuates High Fat Diet-Induced Cerebellar Changes in Rats: Involvement of TNFα, autophagy and oxidative stress. Cells Tissues Organs 2021; 210:351-367. [PMID: 34551416 DOI: 10.1159/000519088] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/12/2021] [Indexed: 11/19/2022] Open
Affiliation(s)
- Hasnaa Ali Ebrahim
- Department of Basic Medical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
- Department of Anatomy, Faculty of Medicine, University of Mansoura, Mansoura, Egypt
| | - Randa El-Gamal
- Department of Medical Biochemistry, Faculty of Medicine, University of Mansoura, Mansoura, Egypt
- Medical Experimental Research Center (MERC), Faculty of Medicine, University of Mansoura, Mansoura, Egypt
| | - Rania N Sherif
- Department of Anatomy, Faculty of Medicine, University of Mansoura, Mansoura, Egypt
- Department of Anatomy and Embryology, Faculty of Dentistry, Horus University, Damietta, Egypt
- Department of Anatomy and Embryology, Faculty of Medicine, Damietta University, Damietta, Egypt
| |
Collapse
|
21
|
Oxidative Stress Enhances Autophagy-Mediated Death Of Stem Cells Through Erk1/2 Signaling Pathway - Implications For Neurotransplantations. Stem Cell Rev Rep 2021; 17:2347-2358. [PMID: 34487308 DOI: 10.1007/s12015-021-10212-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2021] [Indexed: 12/18/2022]
Abstract
Stem cell therapies are becoming increasingly popular solutions for neurological disorders. However, there is a lower survival rate of these cells after transplantation. Oxidative stress is linked to brain damage, and it may also impact transplanted stem cells. To better understand how transplanted cells respond to oxidative stress, the current study used H2O2. We briefly illustrated that exogenous H2O2 treatment exaggerated oxidative stress in the human dental pulp and mesenchymal stem cells. 2',7'-Dichlorofluorescin diacetate (DCFDA), MitoSOX confirms the reactive oxygen species (ROS) involvement, which was remarkably subsided by the ROS inhibitors. The findings showed that H2O2 activates autophagy by enhancing pro-autophagic proteins, Beclin1 and Atg7. Increased LC3II/I expression (which co-localized with lysosomal proteins, LAMP1 and Cathepsin B) showed that H2O2 treatment promoted autophagolysosome formation. In the results, both Beclin1 and Atg7 were observed co-localized in mitochondria, indicating their involvement in mitophagy. The evaluation of Erk1/2 in the presence and absence of Na-Pyruvate, PEG-Catalase, and PD98059 established ROS-Erk1/2 participation in autophagy regulation. Further, these findings showed a link between apoptosis and autophagy. The results conclude that H2O2 acts as a stressor, promoting autophagy and mitophagy in stem cells under oxidative stress. The current study may help understand better cell survival and death approaches for transplanted cells in various neurological diseases. The current study uses human Dental Pulp and Mesenchymal Stem cells to demonstrate the importance of H2O2-driven autophagy in deciding the fate of these cells in an oxidative microenvironment. To summarise, we discovered that exogenous H2O2 treatment causes oxidative stress. Exogenous H2O2 treatment also increased ROS production, especially intracellular H2O2. H2O2 stimulated the ErK1/2 signaling pathway and autophagy. Erk1/2 was found to cause autophagy. Further, the function of mitophagy appeared to be an important factor in the H2O2-induced regulation of these two human stem cell types. In a nutshell, by engaging in autophagy nucleation, maturation, and terminal phase proteins, we elucidated the participation of autophagy in cell dysfunction and death.
Collapse
|
22
|
Crosstalk between Autophagy and Inflammatory Processes in Cancer. Life (Basel) 2021; 11:life11090903. [PMID: 34575052 PMCID: PMC8466094 DOI: 10.3390/life11090903] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/25/2021] [Accepted: 08/28/2021] [Indexed: 12/18/2022] Open
Abstract
Inflammation is an adaptive response to tissue injury, which is a critical process in order to restore tissue functionality and homeostasis. The association between inflammation and cancer has been a topic of interest for many years, not only inflammatory cells themselves but also the chemokines and cytokines they produce, which affect cancer development. Autophagy is an intracellular self-degradative process providing elimination of damaged or dysfunctional organelles under stressful conditions such as nutrient deficiency, hypoxia, or chemotherapy. Interestingly, the signaling pathways that are involved in cancer-associated inflammation may regulate autophagy as well. These are (1) the toll-like receptor (TLR) signaling cascade, (2) the reactive oxygen species (ROS) signaling pathway, (3) the inflammatory cytokine signaling pathway, and (4) the IκB kinase (IKK)/Nuclear factor-κB (NF-κB) signaling axis. Moreover, the studies on the context-specific functions of autophagy during inflammatory responses in cancer will be discussed here. On that basis, we focus on autophagy inhibitors and activators regulating inflammatory process in cancer as useful candidates for enhancing anticancer effects. This review summarizes how the autophagic process regulates these key inflammatory processes and vice versa in various cancers.
Collapse
|
23
|
Role of Herbal Teas in Regulating Cellular Homeostasis and Autophagy and Their Implications in Regulating Overall Health. Nutrients 2021; 13:nu13072162. [PMID: 34201882 PMCID: PMC8308238 DOI: 10.3390/nu13072162] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/19/2021] [Accepted: 06/20/2021] [Indexed: 02/06/2023] Open
Abstract
Tea is one of the most popular and widely consumed beverages worldwide, and possesses numerous potential health benefits. Herbal teas are well-known to contain an abundance of polyphenol antioxidants and other ingredients, thereby implicating protection and treatment against various ailments, and maintaining overall health in humans, although their mechanisms of action have not yet been fully identified. Autophagy is a conserved mechanism present in organisms that maintains basal cellular homeostasis and is essential in mediating the pathogenesis of several diseases, including cancer, type II diabetes, obesity, and Alzheimer’s disease. The increasing prevalence of these diseases, which could be attributed to the imbalance in the level of autophagy, presents a considerable challenge in the healthcare industry. Natural medicine stands as an effective, safe, and economical alternative in balancing autophagy and maintaining homeostasis. Tea is a part of the diet for many people, and it could mediate autophagy as well. Here, we aim to provide an updated overview of popular herbal teas’ health-promoting and disease healing properties and in-depth information on their relation to autophagy and its related signaling molecules. The present review sheds more light on the significance of herbal teas in regulating autophagy, thereby improving overall health.
Collapse
|
24
|
du Plessis M, Davis T, Loos B, Pretorius E, de Villiers WJS, Engelbrecht AM. Molecular regulation of autophagy in a pro-inflammatory tumour microenvironment: New insight into the role of serum amyloid A. Cytokine Growth Factor Rev 2021; 59:71-83. [PMID: 33727011 DOI: 10.1016/j.cytogfr.2021.01.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023]
Abstract
Chronic inflammation, systemic or local, plays a vital role in tumour progression and metastasis. Dysregulation of key physiological processes such as autophagy elicit unfavourable immune responses to induce chronic inflammation. Cytokines, growth factors and acute phase proteins present in the tumour microenvironment regulate inflammatory responses and alter crosstalk between various signalling pathways involved in the progression of cancer. Serum amyloid A (SAA) is a key acute phase protein secreted by the liver during the acute phase response (APR) following infection or injury. However, cancer and cancer-associated cells produce SAA, which when present in high levels in the tumour microenvironment contributes to cancer initiation, progression and metastasis. SAA can activate several signalling pathways such as the PI3K and MAPK pathways, which are also known modulators of the intracellular degradation process, autophagy. Autophagy can be regarded as having a double edged sword effect in cancer. Its dysregulation can induce malignant transformation through metabolic stress which manifests as oxidative stress, endoplasmic reticulum (ER) stress and DNA damage. On the other hand, autophagy can promote cancer survival during metabolic stress, hypoxia and senescence. Autophagy has been utilised to promote the efficiency of chemotherapeutic agents and can either be inhibited or induced to improve treatment outcomes. This review aims to address the known mechanisms that regulate autophagy as well as illustrating the role of SAA in modulating these pathways and its clinical implications for cancer therapy.
Collapse
Affiliation(s)
- M du Plessis
- Department of Physiological Sciences, University of Stellenbosch, Stellenbosch, South Africa.
| | - T Davis
- Department of Physiological Sciences, University of Stellenbosch, Stellenbosch, South Africa
| | - B Loos
- Department of Physiological Sciences, University of Stellenbosch, Stellenbosch, South Africa
| | - E Pretorius
- Department of Physiological Sciences, University of Stellenbosch, Stellenbosch, South Africa
| | - W J S de Villiers
- African Cancer Institute (ACI), Department of Global Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa; Department of Internal Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg Campus, South Africa
| | - A M Engelbrecht
- Department of Physiological Sciences, University of Stellenbosch, Stellenbosch, South Africa; Department of Internal Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg Campus, South Africa
| |
Collapse
|
25
|
Ahmed LA, Abd El-Rhman RH, Gad AM, Hassaneen SK, El-Yamany MF. Dibenzazepine combats acute liver injury in rats via amendments of Notch signaling and activation of autophagy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:337-348. [PMID: 32984915 DOI: 10.1007/s00210-020-01977-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 09/13/2020] [Indexed: 02/08/2023]
Abstract
Paracetamol is a commonly used over-the-counter analgesic and antipyretic drug. Nevertheless, an overdose of paracetamol leads to hepatic necrosis that can be lethal. This study aimed to assess the potential hepatoprotective effects of dibenzazepine, a Notch inhibitor, against acute liver injury in rats via interfering with oxidative stress, inflammation, apoptosis, autophagy, and Notch signaling. Silymarin (200 mg/kg, p.o.) or dibenzazepine (2 mg/kg, i.p.) were administered to rats for 5 days before a single hepatotoxic dose of paracetamol (800 mg/kg, i.p.). Pretreatment with silymarin and dibenzazepine significantly mitigated oxidative stress, inflammatory and apoptotic markers induced by paracetamol hepatotoxicity where dibenzazepine showed greater repression of inflammation. Furthermore, dibenzazepine was found to be significantly more efficacious than silymarin in inhibiting Notch signaling as represented by expression of Notch-1 and Hes-1. A significantly greater response was also demonstrated with dibenzazepine pretreatment with regard to the expression of autophagic proteins, Beclin-1 and LC-3. The aforementioned biochemical results were confirmed by histopathological examination. Autophagy and Notch signaling seem to play a significant role in protection provided by dibenzazepine for paracetamol-induced hepatotoxicity in rats, which could explain its superior results relative to silymarin. Graphical abstract.
Collapse
Affiliation(s)
- Lamiaa A Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Rana H Abd El-Rhman
- Department of Pharmacology, Egyptian Drug Authority formerly National Organization for Drug Control and Research, Giza, Egypt
| | - Amany M Gad
- Department of Pharmacology, Egyptian Drug Authority formerly National Organization for Drug Control and Research, Giza, Egypt
| | - Sherifa K Hassaneen
- Department of Pharmacology, Egyptian Drug Authority formerly National Organization for Drug Control and Research, Giza, Egypt
| | - Mohamad F El-Yamany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
26
|
Li H, Wang R, Yu Z, Shi R, Zhang J, Gao S, Shao M, Cui S, Gao Z, Xu J, Sy MS, Li C. Tumor Necrosis Factor α Reduces SNAP29 Dependent Autolysosome Formation to Increase Prion Protein Level and Promote Tumor Cell Migration. Virol Sin 2020; 36:458-475. [PMID: 33237393 DOI: 10.1007/s12250-020-00320-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/10/2020] [Indexed: 12/20/2022] Open
Abstract
Tumor Necrosis Factor α (TNFα) is best known as a mediator of inflammation and immunity, and also plays important roles in tumor biology. However, the role of TNFα in tumor biology is complex and not completely understood. In a human melanoma cell line, M2, and a lung carcinoma cell line, A549, TNFα up-regulates prion protein (PrP) level, and promotes tumor cell migration in a PrP dependent manner. Silencing PRNP abrogates TNFα induced tumor cell migration; this phenotype is reversed when PRNP is re-introduced. Treatment with TNFα activates nuclear factor kappa B (NF-κB) signaling, which then mitigates autophagy by reducing the expression of Forkhead Box P3 (FOXP3). Down regulation of FOXP3 reduces the transcription of synaptosome associated protein 29 (SNAP29), which is essential in the fusion of autophagosome and lysosome creating autolysosome. FOXP3 being a bona fide transcription factor for SNAP29 is confirmed in a promoter binding assay. Accordingly, silencing SNAP29 in these cell lines also up-regulates PrP, and promotes tumor cell migration without TNFα treatment. But, when SNAP29 or FOXP3 is silenced in these cells, they are no longer respond to TNFα. Thus, a reduction in autophagy is the underlying mechanism by which expression of PrP is up-regulated, and tumor cell migration is enhanced upon TNFα treatment. Disrupting the TNFα-NF-κB-FOXP3-SNAP29 signaling axis may provide a therapeutic approach to mitigate tumor cell migration.
Collapse
Affiliation(s)
- Huan Li
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.,University of Chinese Academy of Sciences, Beijing, 100000, China.,Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 510095, China
| | - Ren Wang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 510095, China
| | - Ze Yu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 510095, China
| | - Run Shi
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 510095, China
| | - Jie Zhang
- Department of Stomatology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832008, China
| | - Shanshan Gao
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Ming Shao
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Shuzhong Cui
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 510095, China.,Abdominal Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Zhenxing Gao
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 510095, China
| | - Jiang Xu
- Department of Stomatology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832008, China
| | - Man-Sun Sy
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Chaoyang Li
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China. .,Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 510095, China.
| |
Collapse
|
27
|
Nouri Z, Fakhri S, Nouri K, Wallace CE, Farzaei MH, Bishayee A. Targeting Multiple Signaling Pathways in Cancer: The Rutin Therapeutic Approach. Cancers (Basel) 2020; 12:E2276. [PMID: 32823876 PMCID: PMC7463935 DOI: 10.3390/cancers12082276] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/07/2020] [Accepted: 08/10/2020] [Indexed: 12/21/2022] Open
Abstract
Multiple dysregulated signaling pathways are implicated in the pathogenesis of cancer. The conventional therapies used in cancer prevention/treatment suffer from low efficacy, considerable toxicity, and high cost. Hence, the discovery and development of novel multi-targeted agents to attenuate the dysregulated signaling in cancer is of great importance. In recent decades, phytochemicals from dietary and medicinal plants have been successfully introduced as alternative anticancer agents due to their ability to modulate numerous oncogenic and oncosuppressive signaling pathways. Rutin (also known as rutoside, quercetin-3-O-rutinoside and sophorin) is an active plant-derived flavonoid that is widely distributed in various vegetables, fruits, and medicinal plants, including asparagus, buckwheat, apricots, apples, cherries, grapes, grapefruit, plums, oranges, and tea. Rutin has been shown to target various inflammatory, apoptotic, autophagic, and angiogenic signaling mediators, including nuclear factor-κB, tumor necrosis factor-α, interleukins, light chain 3/Beclin, B cell lymphoma 2 (Bcl-2), Bcl-2 associated X protein, caspases, and vascular endothelial growth factor. A comprehensive and critical analysis of the anticancer potential of rutin and associated molecular targets amongst various cancer types has not been performed previously. Accordingly, the purpose of this review is to present an up-to-date and critical evaluation of multiple cellular and molecular mechanisms through which the anticancer effects of rutin are known to be exerted. The current challenges and limitations as well as future directions of research are also discussed.
Collapse
Affiliation(s)
- Zeinab Nouri
- Student Research Committee, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah 6714415153, Iran;
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;
| | - Keyvan Nouri
- Student Research Committee, School of Medicine, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran;
| | - Carly E. Wallace
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA;
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA;
| |
Collapse
|
28
|
Thach TT, Wu C, Hwang KY, Lee SJ. Azelaic Acid Induces Mitochondrial Biogenesis in Skeletal Muscle by Activation of Olfactory Receptor 544. Front Physiol 2020; 11:329. [PMID: 32411005 PMCID: PMC7199515 DOI: 10.3389/fphys.2020.00329] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 03/20/2020] [Indexed: 12/31/2022] Open
Abstract
Mouse olfactory receptor 544 (Olfr544) is ectopically expressed in varied extra-nasal organs with tissue specific functions. Here, we investigated the functionality of Olfr544 in skeletal muscle cells and tissue. The expression of Olfr544 is confirmed by RT-PCR and qPCR in skeletal muscle cells and mouse skeletal muscle assessed by RT-PCR and qPCR. Olfr544 activation by its ligand, azelaic acid (AzA, 50 μM), induced mitochondrial biogenesis and autophagy in cultured skeletal myotubes by induction of cyclic adenosine monophosphate-response element binding protein (CREB)-peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α)-extracellular signal-regulated kinase-1/2 (ERK1/2) signaling axis. The silencing Olfr544 gene expression abrogated these effects of AzA in cultured myotubes. Similarly, in mice, the acute subcutaneous injection of AzA induced the CREB-PGC-1α-ERK1/2 pathways in mouse skeletal muscle, but these activations were negated in those of Olfr544 knockout mice. These demonstrate that the induction of mitochondrial biogenesis in skeletal muscle by AzA is Olfr544-dependent. Oral administration of AzA to high-fat-diet fed obese mice for 6 weeks increased mitochondrial DNA content in the skeletal muscle as well. Collectively, these findings demonstrate that Olfr544 activation by AzA regulates mitochondrial biogenesis in skeletal muscle. Intake of AzA or food containing AzA may help to improve skeletal muscle function.
Collapse
Affiliation(s)
- Trung Thanh Thach
- Department of Biotechnology, School of Life Sciences and Biotechnology for BK21-PLUS, Korea University, Seoul, South Korea.,Department of Food Bioscience and Technology, College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| | - Chunyan Wu
- Department of Biotechnology, School of Life Sciences and Biotechnology for BK21-PLUS, Korea University, Seoul, South Korea.,Department of Food Bioscience and Technology, College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| | - Kwang Yeon Hwang
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| | - Sung-Joon Lee
- Department of Biotechnology, School of Life Sciences and Biotechnology for BK21-PLUS, Korea University, Seoul, South Korea.,Department of Food Bioscience and Technology, College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| |
Collapse
|
29
|
Xiang H, Zhang J, Lin C, Zhang L, Liu B, Ouyang L. Targeting autophagy-related protein kinases for potential therapeutic purpose. Acta Pharm Sin B 2020; 10:569-581. [PMID: 32322463 PMCID: PMC7161711 DOI: 10.1016/j.apsb.2019.10.003] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/06/2019] [Accepted: 09/09/2019] [Indexed: 02/08/2023] Open
Abstract
Autophagy, defined as a scavenging process of protein aggregates and damaged organelles mediated by lysosomes, plays a significant role in the quality control of macromolecules and organelles. Since protein kinases are integral to the autophagy process, it is critically important to understand the role of kinases in autophagic regulation. At present, intervention of autophagic processes by small-molecule modulators targeting specific kinases has becoming a reasonable and prevalent strategy for treating several varieties of human disease, especially cancer. In this review, we describe the role of some autophagy-related kinase targets and kinase-mediated phosphorylation mechanisms in autophagy regulation. We also summarize the small-molecule kinase inhibitors/activators of these targets, highlighting the opportunities of these new therapeutic agents.
Collapse
Key Words
- 4E-BP1, eukaryotic translation initiation factor 4E-binding protein
- AKT1, AKT serine/threonine kinase 1
- AMBRA1, autophagy/beclin-1 regulator 1
- AMPK, AMP-activated protein kinase
- ARF, auxin response factor gene
- ATG, autophagy-related protein
- Autophagy
- Autophagy-related kinase
- CaMKK2, calcium/calmodulin-dependent protein kinase kinase 2
- DAPK, death associated protein kinase
- FIP200, FAK family kinase-interacting protein of 200 kDa
- GAP, GTPase-activating protein
- GO, gene ontology
- GSK3α, glycogen synthase kinase 3 alpha
- HMGB1, high mobility group protein B1
- Human disease therapy
- JNK1, C-Jun N-terminal kinase
- LC3, microtubule-associated protein 1 light chain 3
- LKB1, serine/threonine-protein kinase stk11
- LPS, lipopolysaccharide
- LRRK2, leucine rich repeat kinase 2
- PD, Parkinson's disease
- PI, phosphatidylinositol
- PI3 kinase, phosphoinositide 3-kinase
- PI3P, phosphatidylinositol triphosphate
- PIM2, proviral insertion in murine lymphomas 2
- PINK1, PTEN-induced putative kinase 1
- PIP2, phosphatidylinositol-4,5-bisphosphate
- PKACα, a protein kinase cAMP-activated catalytic subunit alpha
- PKCα, protein kinase C alpha type
- PKD1, polycystin-1
- PPIs, protein–protein interactions
- PROTAC, proteolysis targeting chimeras
- PTMs, post-translational modifications
- Phosphorylation
- Protein kinases
- Rheb, the RAS homolog enriched in brain
- Small-molecule kinase inhibitors/activators
- TAK1, transforming growth factor activated kinase-1
- TFEB, transcription factor EB
- TNBC, triple-negative breast cancer
- TSC1/2, tuberous sclerosis complex proteins 1/2
- ULK complex, ULK1–mATG13–FIP200–ATG101 complex
- ULK1, unc-51-like kinase 1
- UVRAG, ultraviolet resistance-associated gene
- mTOR, mammalian target of rapamycin
- mTORC1, mammalian target of rapamycin complex 1
Collapse
Affiliation(s)
- Honggang Xiang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Jifa Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Congcong Lin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Lan Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, China
| |
Collapse
|
30
|
Nerome K, Ito-Kureha T, Paganini T, Fukuda T, Igarashi Y, Ashitomi H, Ikematsu S, Yamamoto T. Potent and broad anticancer activities of leaf extracts from Melia azedarach L. of the subtropical Okinawa islands. Am J Cancer Res 2020; 10:581-594. [PMID: 32195029 PMCID: PMC7061759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 01/23/2020] [Indexed: 06/10/2023] Open
Abstract
Plant extracts have been traditionally used for various therapeutic applications. By conducting an initial screening of several subtropical plants, in this study, we evaluated the anticancer activities of Melia azedarach L. The extract from Melia azedarach L. leaves (MLE) show high cytotoxic effects on cancer cells and in vivo mouse and dog tumor models. During the initial screening, MLE showed strong antiproliferative activity against HT-29 colon, A549 lung, and MKN1 gastric cancer cells. In subsequent tests, using 39 human tumor cell lines, we confirmed the potent anticancer activities of MLE. The anticancer activity of MLE was also confirmed in vivo. MLE markedly inhibited the growth of transplanted gastric MKN1 cancer xenografts in mice. To elucidate the mechanism underlying the anticancer effects of MLE, MLE-treated MKN1 cells were observed using an electron microscope; MLE treatment induced autophagy. Furthermore, western blot analysis of proteins in lysates of MLE-treated cells revealed induction of light chain 3 (LC3)-II autophagosomal proteins. Thus, MLE appeared to suppress MKN1 cell proliferation by inducing autophagy. In addition, in the mouse macrophage cell line J774A.1, MLE treatment induced TNF-α production, which might play a role in tumor growth suppression in vivo. We also performed a preclinical evaluation of MLE treatment on dogs with various cancers in veterinary hospitals. Dogs with various types of cancers showed a mean recovery of 76% when treated with MLE. Finally, we tried to identify the active substances present in MLE. All the active fractions obtained by reverse-phase chromatography contained azedarachin B-related moieties, such as 3-deacetyl-12-hydroxy-amoorastatin, 12-hydroxy-amoorastatin, and 12-hydroxyamoorastaton. In conclusion, MLE contains substances with promising anticancer effects, suggesting their future use as safe and effective anticancer agents.
Collapse
Affiliation(s)
- Kuniaki Nerome
- The Institute of Biological Resources893-2, Nakayama, Nago-shi, Okinawa 905-0004, Japan
| | - Taku Ito-Kureha
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate UniversityOnna, Okinawa 904-0495, Japan
| | - Tiziana Paganini
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate UniversityOnna, Okinawa 904-0495, Japan
| | - Takao Fukuda
- Biotechnology Research Center and Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University5180 Kurokawa, Imizu, Toyama 939-0398, Japan
| | - Yasuhiro Igarashi
- Biotechnology Research Center and Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University5180 Kurokawa, Imizu, Toyama 939-0398, Japan
| | - Hiroto Ashitomi
- National Institute of Technology, Okinawa CollegeHenoko 905, Nago-shi, Okinawa 905-2192, Japan
| | - Shinya Ikematsu
- National Institute of Technology, Okinawa CollegeHenoko 905, Nago-shi, Okinawa 905-2192, Japan
| | - Tadashi Yamamoto
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate UniversityOnna, Okinawa 904-0495, Japan
| |
Collapse
|
31
|
Li X, He S, Ma B. Autophagy and autophagy-related proteins in cancer. Mol Cancer 2020; 19:12. [PMID: 31969156 PMCID: PMC6975070 DOI: 10.1186/s12943-020-1138-4] [Citation(s) in RCA: 1030] [Impact Index Per Article: 206.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 01/16/2020] [Indexed: 12/19/2022] Open
Abstract
Autophagy, as a type II programmed cell death, plays crucial roles with autophagy-related (ATG) proteins in cancer. Up to now, the dual role of autophagy both in cancer progression and inhibition remains controversial, in which the numerous ATG proteins and their core complexes including ULK1/2 kinase core complex, autophagy-specific class III PI3K complex, ATG9A trafficking system, ATG12 and LC3 ubiquitin-like conjugation systems, give multiple activities of autophagy pathway and are involved in autophagy initiation, nucleation, elongation, maturation, fusion and degradation. Autophagy plays a dynamic tumor-suppressive or tumor-promoting role in different contexts and stages of cancer development. In the early tumorigenesis, autophagy, as a survival pathway and quality-control mechanism, prevents tumor initiation and suppresses cancer progression. Once the tumors progress to late stage and are established and subjected to the environmental stresses, autophagy, as a dynamic degradation and recycling system, contributes to the survival and growth of the established tumors and promotes aggressiveness of the cancers by facilitating metastasis. This indicates that regulation of autophagy can be used as effective interventional strategies for cancer therapy.
Collapse
Affiliation(s)
- Xiaohua Li
- Henan Provincial People's Hospital, Zhengzhou, 450003, China.,Henan Eye Hospital, Henan Eye Institute, Henan Key Laboratory of Ophthalmology and Visual Science, Zhengzhou, 450003, China.,People's Hospital of Zhengzhou University, Zhengzhou, 450003, China.,People's Hospital of Henan University, Zhengzhou, 450003, China
| | - Shikun He
- Ophthalmology Optometry Centre, Peking University People's Hospital, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, 100044, China.,Department of Pathology and Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, 90033, USA
| | - Binyun Ma
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, 90033, USA. .,Department of Medicine/Hematology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
32
|
Chen J, Che L, Xu C, Zhao S, Yang J, Li M, Li G, Shen Y. Cardio-facio-cutaneous syndrome-associated pathogenic MAP2K1 variants activate autophagy. Gene 2020; 733:144369. [PMID: 31972311 DOI: 10.1016/j.gene.2020.144369] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/12/2020] [Accepted: 01/13/2020] [Indexed: 02/08/2023]
Abstract
MAP2K1 encodes mitogen-activated protein kinase 1 (MEK1). Mutations in MAP2K1 lead to continuous activation of MEK/ERK signaling pathway, giving rise to cardio-facio-cutaneous syndrome (CFCS). However, the molecular mechanisms of abnormal activation of MEK/ERK signaling pathway and the role of autophagy, if any, in manifesting CFCS in MAP2K mutants remain unclear. Here, we report three Chinese children with CFCS having MAP2K1 pathogenic variants, identified by exome sequencing. They presented with dysmorphic facial features, seizures, psychomotor retardation, and short stature. Additionally, the third child showed pulmonary valve stenosis, multiple skeletal deformities, and osteoporosis. Whole exome sequencing revealed two heterozygous missense mutations in exon 3 of MAP2K1 (c.383G>T; p.Gly128Val and c.389A>G; p.Tyr130Cys), as well as a novel heterozygous missense variant (c.170A>T; p.Lys57Met) in exon 2 of MAP2K1. In SH-SY5Y cells, we identified, for the first time, that MAP2K1 mutations can activate the p-ERK-dependent cell cycle progression and autophagy, and cause CFCS. Our results extended the mutational spectrum of MAP2K1, examined the role of MEK1 protein in nerve cell functions, and demonstrated, for the first time, that autophagy may mediate the altered MAP2K1 function, leading to CFCS phenotypes.
Collapse
Affiliation(s)
- Jing Chen
- Department of Child Health, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China; School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Lin Che
- School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Chao Xu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, China; Shandong Clinical Medical Center of Endocrinology and Metabolism, Jinan, China
| | - Suzhou Zhao
- Fulgent Technologies Inc, Minhang District, Shanghai, China
| | - Jiangfei Yang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Mengting Li
- Genetic and Metabolic Central Laboratory, Birth Defect Prevention Research Institute, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Guimei Li
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.
| | - Yiping Shen
- Genetic and Metabolic Central Laboratory, Birth Defect Prevention Research Institute, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China; Division of Genetics and Genomics, Boston Children's Hospital, Department of Neurology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
33
|
Nuñez-Olvera SI, Gallardo-Rincón D, Puente-Rivera J, Salinas-Vera YM, Marchat LA, Morales-Villegas R, López-Camarillo C. Autophagy Machinery as a Promising Therapeutic Target in Endometrial Cancer. Front Oncol 2019; 9:1326. [PMID: 31850214 PMCID: PMC6896250 DOI: 10.3389/fonc.2019.01326] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 11/13/2019] [Indexed: 01/01/2023] Open
Abstract
Endometrial cancer is the fourth most frequent neoplasia for women worldwide, and over the past two decades it incidence has increased. The most common histological type of endometrial cancer is endometrioid adenocarcinoma, also known as type 1 endometrial cancer. Endometrioid endometrial cancer is associated with diverse epidemiological risk factors including estrogen use, obesity, diabetes, cigarette smoking, null parity, early menarche, and late menopause. Clinical effectiveness of chemotherapy is variable, indicating that novel molecular therapies against specific cellular processes associated to cell survival and resistance to therapy, such as autophagy, urged to ameliorate the rates of success in endometrial cancer treatment. Autophagy (also known as macroautophagy) is a specialized mechanism that maintains cell homeostasis which is activated in response to cellular stressors including nutrients deprivation, amino acids starvation, hypoxia, and metabolic stress to prolong cell survival via lysosomal degradation of cytoplasmic macromolecules and organelles. However, in human cancer cells, autophagy has a controversial function due to its dual role as self-protective or apoptotic. Conventional antitumor therapies including hormones, chemotherapy and ionizing radiation, may activate autophagy as a pro-survival tumor response contributing to treatment resistance. Intriguingly, if autophagy continues above reversibility of cell viability, autophagy can result in apoptosis of tumor cells. Here, we have reviewed the mechanisms of autophagy described in endometrial cancers, including the role of PI3K/AKT/mTOR, AMPK-mTOR, and p53 signaling pathways that trigger or inhibit the process and thus representing potential molecular targets in therapeutic clinical approaches. In addition, we discussed the recent findings indicating that autophagy can be modulated using repurposing drugs which may leads to faster experimentation and validation, as well as more easy access of the medications to patients. Finally, the promising role of dietary compounds and microRNAs in autophagy modulation is also discussed. In conclusion, although the research about autophagy is scarce but ongoing in endometrial cancer, the actual findings highlight the promising usefulness of novel molecules for directing targeted therapies.
Collapse
Affiliation(s)
| | - Dolores Gallardo-Rincón
- Laboratorio de Medicina Translacional, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Jonathan Puente-Rivera
- Departamento de Ecología Funcional, Instituto de Ecología, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Yarely M. Salinas-Vera
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Mexico City, Mexico
| | - Laurence A. Marchat
- Programa en Biomedicina Molecular y Red de Biotecnología, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Raúl Morales-Villegas
- Coordinación Académica Huasteca del Sur, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Mexico City, Mexico
| |
Collapse
|
34
|
Yang X, Zheng YT, Rong W. Sevoflurane induces apoptosis and inhibits the growth and motility of colon cancer in vitro and in vivo via inactivating Ras/Raf/MEK/ERK signaling. Life Sci 2019; 239:116916. [PMID: 31626792 DOI: 10.1016/j.lfs.2019.116916] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 09/17/2019] [Accepted: 09/25/2019] [Indexed: 12/21/2022]
Abstract
AIMS To investigate the effects of sevoflurane on proliferation, cell cycle, apoptosis, autophagy, invasion and epithelial-mesenchymal transition of colon cancer cell line SW480, and to explore its possible mechanism. MATERIALS AND METHODS SW480 and SW620 cells were treated with a mixture of 95% O2+5% CO2 containing different concentrations of sevoflurane (1.7% SAV, 3.4% SAV and 5.1% SAV) for 6 h. Meanwhile, we performed a rescue experiment by treating cells with the ERK pathway activator LM22B-10 prior to treatment of cells with 5.1% sevoflurane。 KEY FINDINGS: High concentration (5.1%) of sevoflurane significantly inhibited the proliferation and invasion of cells, causing G0/G1 phase arrest and promoted apoptosis and autophagy. 5.1% sevoflurane can participate in the regulation of EMT by regulating the expression of E-cadherin, Vimentin and N-cadherin proteins. LM22B-10 promoted proliferation and invasion of cancer cells and inhibited apoptosis and autophagy, while 5.1% sevoflurane could reverse the effect of LM22B-10 on the biological characteristics of cells. Sevoflurane can significantly inhibit tumor growth in SW480 cells transplanted nude mice. Moreover, 5.1% sevoflurane significantly increased the expression of p-Raf, p-MEK1/2, and p-ERK1/2 in SW480 cells and tumor tissues without affecting p-JNK and p-p38 proteins, meanwhile, 5.1% sevoflurane can inhibit the activation of ERK signaling pathway by LM22B-10 in vitro and in vivo. SIGNIFICANCE Sevoflurane can inhibit the proliferation and invasion of colon cancer cells, induce apoptosis and autophagy, and participate in the regulation of epithelial-mesenchymal transition, which may be related to its inhibition of the ERK signaling pathway.
Collapse
Affiliation(s)
- Xiao Yang
- Department of Anesthesiology, Weihai Central Hospital, Weihai, 264400, Shandong, China
| | - Yao-Tun Zheng
- Department of Anesthesiology, Weihai Central Hospital, Weihai, 264400, Shandong, China
| | - Wei Rong
- Department of Anesthesiology, Weihai Central Hospital, Weihai, 264400, Shandong, China.
| |
Collapse
|
35
|
Subkamkaew C, Limtrakul (Dejkriengkraikul) P, Yodkeeree S. Proanthocyanidin-Rich Fractions from Red Rice Extract Enhance TNF-α-Induced Cell Death and Suppress Invasion of Human Lung Adenocarcinoma Cell A549. Molecules 2019; 24:E3393. [PMID: 31540489 PMCID: PMC6767152 DOI: 10.3390/molecules24183393] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 09/10/2019] [Accepted: 09/17/2019] [Indexed: 01/08/2023] Open
Abstract
Tumor necrosis factor-alpha (TNF-α) plays a key role in promoting tumor progression, such as stimulation of cell proliferation and metastasis via activation of NF-κB and AP-1. The proanthocyanidin-rich fraction obtained from red rice (PRFR) has been reported for its anti-tumor effects in cancer cells. This study investigated the molecular mechanisms associated with PRFR on cell survival and metastasis of TNF-α-induced A549 human lung adenocarcinoma. Notably, PRFR enhanced TNF-α-induced A549 cell death when compared with PRFP alone and caused a G0-G1 cell cycle arrest. Although, PRFR alone enhanced cell apoptosis, the combination treatment induced the cells that had been enhanced with PRFR and TNF-α to apoptosis that was less than PRFR alone and displayed a partial effect on caspase-8 activation and PARP cleavage. By using the autophagy inhibitor; 3-MA attenuated the effect of how PRFR enhanced TNF-α-induced cell death. This indicates that PRFR not only enhanced TNF-α-induced A549 cell death by apoptotic pathway, but also by induction autophagy. Moreover, PRFR also inhibited TNF-α-induced A549 cell invasion. This effect was associated with PRFR suppressed the TNF-α-induced level of expression for survival, proliferation, and invasive proteins. This was due to reduce of MAPKs, Akt, NF-κB, and AP-1 activation. Taken together, our results suggest that TNF-α-induced A549 cell survival and invasion are attenuated by PRFR through the suppression of the MAPKs, Akt, AP-1, and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Chayaporn Subkamkaew
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (C.S.); )
| | - Pornngarm Limtrakul (Dejkriengkraikul)
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (C.S.); )
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Supachai Yodkeeree
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (C.S.); )
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
36
|
Kong T, Liu M, Ji B, Bai B, Cheng B, Wang C. Role of the Extracellular Signal-Regulated Kinase 1/2 Signaling Pathway in Ischemia-Reperfusion Injury. Front Physiol 2019; 10:1038. [PMID: 31474876 PMCID: PMC6702336 DOI: 10.3389/fphys.2019.01038] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 07/29/2019] [Indexed: 12/11/2022] Open
Abstract
Extracellular signal-regulated kinase 1/2 (ERK1/2), an important member of the mitogen-activated protein kinase family, is found in many organisms, and it participates in intracellular signal transduction. Various stimuli induce phosphorylation of ERK1/2 in vivo and in vitro. Phosphorylated ERK1/2 moves to the nucleus, activates many transcription factors, regulates gene expression, and controls various physiological processes, finally inducing repair processes or cell death. With the aging of the population around the world, the occurrence of ischemia-reperfusion injury (IRI), especially in the brain, heart, kidney, and other important organs, is becoming increasingly serious. Abnormal activation of the ERK1/2 signaling pathway is closely related to the development and the metabolic mechanisms of IRI. However, the effects of this signaling pathway and the underlying mechanism differ between various models of IRI. This review summarizes the ERK1/2 signaling pathway and the molecular mechanism underlying its role in models of IRI in the brain, heart, liver, kidneys, and other organs. This information will help to deepen the understanding of ERK1/2 signals and deepen the exploration of IRI treatment based on the ERK1/2 study.
Collapse
Affiliation(s)
- Tingting Kong
- Cheeloo College of Medicine, Shandong University, Jinan, China.,School of Mental Health, Neurobiology Key Laboratory of Jining Medical University in Colleges of Shandong, Jining Medical University, Jining, China
| | - Minghui Liu
- School of Mental Health, Neurobiology Key Laboratory of Jining Medical University in Colleges of Shandong, Jining Medical University, Jining, China
| | - Bingyuan Ji
- School of Mental Health, Neurobiology Key Laboratory of Jining Medical University in Colleges of Shandong, Jining Medical University, Jining, China
| | - Bo Bai
- School of Mental Health, Neurobiology Key Laboratory of Jining Medical University in Colleges of Shandong, Jining Medical University, Jining, China
| | - Baohua Cheng
- School of Mental Health, Neurobiology Key Laboratory of Jining Medical University in Colleges of Shandong, Jining Medical University, Jining, China
| | - Chunmei Wang
- School of Mental Health, Neurobiology Key Laboratory of Jining Medical University in Colleges of Shandong, Jining Medical University, Jining, China
| |
Collapse
|
37
|
Gao Y, Qi W, Liu S, Zhao S, Lv J, Qiu W. Acid-induced autophagy protects human gastric cancer cells from apoptosis by activating Erk1/2 pathway. Transl Cancer Res 2019; 8:1560-1570. [PMID: 35116899 PMCID: PMC8798117 DOI: 10.21037/tcr.2019.07.42] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 05/28/2019] [Indexed: 11/23/2022]
Abstract
Background Acidic microenvironments exist widely in tumors. However, the specific mechanism of cancer cell survival under an acidic microenvironment remains unknown. This study aims to investigate whether acid can induce autophagy and examine the mechanism of autophagy in gastric cancer cells. Methods Human gastric adenocarcinoma (AGS) cells were cultured in media with different pH values in vitro and then subjected to autophagy detection under different conditions. To determine the effect of an acidic microenvironment on autophagy, we employed real-time quantitative polymerase chain reaction (PCR), Western blot, mRFP-GFP-LC3 immunofluorescence, and transmission electron microscopy (TEM) to detect the expression of various autophagy indicators. We also performed cell counting kit 8 (CCK8) and cell invasion and migration assays to examine cell viability and invasion, respectively. Results We found that the protein expression of autophagy markers such as LC3II/I and Beclin1 was higher in AGS cells treated with an acidic microenvironment than in control cells. The protein expression level of P62 was obviously decreased in acid-treated cells compared to that in control cells. Furthermore, the expression of Erk1/2 pathway markers, including p-Erk1/2, was also increased in response to acidic pH. Dense LC3 puncta were observed in cells cultured under acidic conditions, whereas untreated cells exhibited diffuse and weak LC3 puncta; an increased autophagy flux could also be observed. The presence of autophagosomes was observed by TEM in AGS cells subjected to low pH. Additionally, autophagy was inhibited by the autophagy inhibitor Bafilomycin A1 (Baf) and apoptosis was obviously increased. Moreover, cells exposed to an acidic microenvironment displayed facilitated growth compared with that in control cells. Conclusions Taken together, these results indicate that the acidic microenvironment promotes AGS cell growth by upregulating autophagy through the Erk1/2 pathway, which acts as a survival adaptation.
Collapse
Affiliation(s)
- Yuan Gao
- Department of Oncology, Qingdao University, Qingdao 266000, China
| | - Weiwei Qi
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Shihai Liu
- Central Laboratory, Affiliated Hospital of Qingdao University School, Qingdao 266000, China
| | - Shufen Zhao
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Jing Lv
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Wensheng Qiu
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao 266000, China
| |
Collapse
|
38
|
Yang S, Yang L, Li X, Li B, Li Y, Zhang X, Ma Y, Peng X, Jin H, Li H. New insights into autophagy in hepatocellular carcinoma: mechanisms and therapeutic strategies. Am J Cancer Res 2019; 9:1329-1353. [PMID: 31392073 PMCID: PMC6682711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 06/13/2019] [Indexed: 06/10/2023] Open
Abstract
Autophagy is a mechanism by which cellular substances are transported to lysosomes for degradation, allowing the basic transformation of cellular components, and providing energy and macromolecular precursors. In cancer, the contradictory role of autophagy in tumor suppression and promotion has been widely acknowledged. Activation and suppression of autophagy have been proposed as cancer therapies, resulting in targeted treatment of cancer by autophagy being considered ambiguous. The dynamic effect of autophagy can also be applied to hepatocellular carcinoma (HCC), a malignant tumor with high incidence and a low survival rate. In this review, we introduce characteristics of different types of autophagy and summarize which genes, non-coding RNAs, and related signaling pathways are involved in autophagy and the regulation of the formation and progress of HCC. More importantly, we discuss the role of autophagy in the treatment of HCC, such as in traditional chemotherapy, molecular targeted drugs, and natural products.
Collapse
Affiliation(s)
- Shuo Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Xinyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Bowen Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Yan Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Xiaodong Zhang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Yingbo Ma
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Hongyuan Jin
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| |
Collapse
|
39
|
Derrone induces autophagic cell death through induction of ROS and ERK in A549 cells. PLoS One 2019; 14:e0218659. [PMID: 31216334 PMCID: PMC6583947 DOI: 10.1371/journal.pone.0218659] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 06/06/2019] [Indexed: 01/07/2023] Open
Abstract
We studied the effect of derrone (DR), one of the major compounds of unripe fruits of Cudrania tricuspidata, on cancer cell death. DR inhibited cell growth of various cancer cells, and that was partially associated with apoptosis in A549 cells. DR showed the autophagic features, such as the conversion of LC3-I to LC3-II, the formation of autophagosome and the downregulation of SQSTM1/p62 (p62). The treatment of autophagy inhibitor reversed DR-mediated cell death, suggesting that DR induces autophagic cell death. The increase of cytoplasmic Ca2+ and ROS by DR treatment significantly influences the formation of autophagosomes; however, only ROS scavengers significantly rescued the reduced cell viability. Additional results revealed that treatment of DR induces sustained phosphorylation of ERK and the inhibition of ERK phosphorylation using U0126 (ERK inhibitor) markedly attenuated DR-induced cell death. Overall, these results suggest that DR induces autophagic cell death through intracellular ROS and sustained ERK phosphorylation in A549 cells.
Collapse
|
40
|
Fung TS, Liu DX. The ER stress sensor IRE1 and MAP kinase ERK modulate autophagy induction in cells infected with coronavirus infectious bronchitis virus. Virology 2019; 533:34-44. [PMID: 31082732 PMCID: PMC7112053 DOI: 10.1016/j.virol.2019.05.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/01/2019] [Accepted: 05/01/2019] [Indexed: 12/11/2022]
Abstract
Coronavirus infection induces the generation of autophagosomes, and certain host proteins regulating cellular autophagy are hijacked by some coronaviruses to facilitate the formation of double membrane vesicles. However, mechanisms underlying coronavirus-induced autophagy remain largely unknown. In this study, we demonstrate that autophagosome formation and apparent autophagic flux are induced in cells infected with infectious bronchitis virus (IBV) - a gammacoronavirus. Notably, IBV-induced autophagy was dependent on autophagy related 5 (ATG5) but not beclin1 (BECN1), although both are essential proteins in the canonical autophagy pathway. Moreover, the ER stress sensor inositol requiring enzyme 1 (IRE1), but not its substrate X-box protein 1 (XBP1), was also essential for the induction of autophagy during IBV infection. Finally, the anti-apoptotic extracellular signal-regulated kinase 1/2 (ERK1/2) also contributed to IBV-induced autophagy. Our findings add new knowledge to the regulatory mechanisms governing coronavirus-induced autophagy, highlighting an extensive cross-talk among cellular signaling pathways during coronavirus infection.
Collapse
Affiliation(s)
- To Sing Fung
- South China Agricultural University, Guangdong Province Key Laboratory Microbial Signals & Disease Co, and Integrative Microbiology Research Centre, Guangzhou, 510642, Guangdong, People's Republic of China
| | - Ding Xiang Liu
- South China Agricultural University, Guangdong Province Key Laboratory Microbial Signals & Disease Co, and Integrative Microbiology Research Centre, Guangzhou, 510642, Guangdong, People's Republic of China.
| |
Collapse
|
41
|
NF-κB/mTOR-mediated autophagy can regulate diquat-induced apoptosis. Arch Toxicol 2019; 93:1239-1253. [PMID: 30848314 DOI: 10.1007/s00204-019-02424-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 03/04/2019] [Indexed: 12/19/2022]
Abstract
Autophagy and apoptosis are the major types of cell death in pesticide-induced neurotoxicity, and autophagy is known to play a role in cell protection by inhibiting apoptosis. In this study, we characterized the relationship between autophagy and apoptosis in diquat (DQ)-induced cell death and explored a novel pharmacotherapeutic approach involving autophagy regulation to prevent DQ neurotoxicity. DQ was cytotoxic to PC12 cells in a concentration-dependent manner, as shown by decreased cell viability and decreased dopamine (DA) levels. DQ-induced apoptosis was found in PC12 cells, as demonstrated by activation of caspase-3 and -9 and by nuclear condensation. By monitoring expression of microtubule-associated protein 1A/1B light chain 3B (LC3-II) and p62, DQ was found to induce autophagy. Exposure of PC12 cells to DQ led to the production of reactive oxygen species (ROS), and N-acetyl-cysteine (NAC) antioxidant effectively blocked both apoptosis and autophagy. Interestingly, DQ in PC12 cells showed increased p53 and NF-κB in a time-dependent manner; furthermore, pifithrin-α (PFT-α), a p53 inhibitor, downregulates the cytotoxicity of DQ, as shown by decreased LC3-II and cleaved caspase-3. SN50, an NF-κB inhibitor, results in diminished LC3-II, cleaved caspase-3, and p53. DQ induces mitogen-activated protein kinase (MAPK) signaling including ERK, JNK, and p38, which inhibit regulated apoptosis and autophagic cell death by controlling mTOR signaling. In addition, modulation of DQ-induced apoptosis in response to autophagy regulation was investigated. Pretreatment with rapamycin, an autophagy inducer, significantly enhanced the viability of DQ-exposed cells by alleviating DQ-induced apoptosis. Conversely, cell pretreatment with 3-methyladenine (3MA), an autophagy inhibitor increased DQ toxicity. Our results suggest that DQ-induced cytotoxicity is modified by autophagy regulation. Pharmacologic induction of autophagy may be a useful treatment strategy in neurodegenerative disorders.
Collapse
|
42
|
Wu D, Zhou WY, Lin XT, Fang L, Xie CM. Bufalin induces apoptosis via mitochondrial ROS-mediated caspase-3 activation in HCT-116 and SW620 human colon cancer cells. Drug Chem Toxicol 2019; 42:444-450. [PMID: 30777466 DOI: 10.1080/01480545.2018.1512611] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Bufalin has been reported to kill various types of cancer including human colorectal cancer. Our previous study demonstrated that bufalin induced cell death via autophagy in HT-29 and Caco-2 colon cancer cells, but the action of bufalin remains unclear. This study was conducted to investigate the role of bufalin in other colon cancer HCT-116 and SW620 cells as well as its potential mechanism. METHODS The effect of bufalin in HCT-116 and SW620 colon cancer cells was detected by assessing cell viability and cell death. Apoptotic cells were analyzed by Western blot and trypan blue dye exclusion assay. Mitochondrial ROS production was analyzed by flow cytometry after DCFDA and DHR-123 staining. The potential mechanism was investigated via pharmacological inhibitors. RESULTS Bufalin had high potency against HCT-116 and SW620 cells with IC50 values of 12.823 ± 1.792 nM and 26.303 ± 2.498 nM in HCT-116 and SW620 cells, respectively. Bufalin decreased cell viability, increased cell death as well as caspase-3 downstream target (cleaved PARP) accumulation, and these actions were significantly blocked by pan-caspase inhibitor zVAD-FMK. Mechanistically, ROS production, but neither the NAD(P)H oxidase, AMPK, ERK nor p38, is responsible for bufalin-induced apoptotic cell death. Moreover, bufalin-induced ROS generation is derived from mitochondria. CONCLUSION Bufalin significantly induces apoptosis in HCT-116 and SW620 colon cancer cells via mitochondrial ROS-mediated caspase-3 activation. We believe that our novel findings will greatly alter our current understanding on the anti-cancer mechanism of bufalin in colon cancer cells and will pave the way for further exploiting the clinical application.
Collapse
Affiliation(s)
- Di Wu
- a Institute of Hepatobiliary Surgery, Southwest Hospital , Third Military Medical University (Army Medical University) , Chongqing , China
| | - Wen-Yi Zhou
- b Essence Securities Co., Ltd. , Chongqing , China
| | - Xiao-Tong Lin
- a Institute of Hepatobiliary Surgery, Southwest Hospital , Third Military Medical University (Army Medical University) , Chongqing , China
| | - Lei Fang
- a Institute of Hepatobiliary Surgery, Southwest Hospital , Third Military Medical University (Army Medical University) , Chongqing , China
| | - Chuan-Ming Xie
- a Institute of Hepatobiliary Surgery, Southwest Hospital , Third Military Medical University (Army Medical University) , Chongqing , China
| |
Collapse
|
43
|
Giampieri F, Afrin S, Forbes-Hernandez TY, Gasparrini M, Cianciosi D, Reboredo-Rodriguez P, Varela-Lopez A, Quiles JL, Battino M. Autophagy in Human Health and Disease: Novel Therapeutic Opportunities. Antioxid Redox Signal 2019; 30:577-634. [PMID: 29943652 DOI: 10.1089/ars.2017.7234] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE In eukaryotes, autophagy represents a highly evolutionary conserved process, through which macromolecules and cytoplasmic material are degraded into lysosomes and recycled for biosynthetic or energetic purposes. Dysfunction of the autophagic process has been associated with the onset and development of many human chronic pathologies, such as cardiovascular, metabolic, and neurodegenerative diseases as well as cancer. Recent Advances: Currently, comprehensive research is being carried out to discover new therapeutic agents that are able to modulate the autophagic process in vivo. Recent evidence has shown that a large number of natural bioactive compounds are involved in the regulation of autophagy by modulating several transcriptional factors and signaling pathways. CRITICAL ISSUES Critical issues that deserve particular attention are the inadequate understanding of the complex role of autophagy in disease pathogenesis, the limited availability of therapeutic drugs, and the lack of clinical trials. In this context, the effects that natural bioactive compounds exert on autophagic modulation should be clearly highlighted, since they depend on the type and stage of the pathological conditions of diseases. FUTURE DIRECTIONS Research efforts should now focus on understanding the survival-supporting and death-promoting roles of autophagy, how natural compounds interact exactly with the autophagic targets so as to induce or inhibit autophagy and on the evaluation of their pharmacological effects in a more in-depth and mechanistic way. In addition, clinical studies on autophagy-inducing natural products are strongly encouraged, also to highlight some fundamental aspects, such as the dose, the duration, and the possible synergistic action of these compounds with conventional therapy.
Collapse
Affiliation(s)
- Francesca Giampieri
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy
| | - Sadia Afrin
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy
| | - Tamara Y Forbes-Hernandez
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy .,2 Area de Nutricion y Salud, Universidad Internacional Iberoamericana , Campeche, Mexico
| | - Massimiliano Gasparrini
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy
| | - Danila Cianciosi
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy
| | - Patricia Reboredo-Rodriguez
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy .,3 Departamento de Quimica Analıtica y Alimentaria, Grupo de Nutricion y Bromatologıa, Universidade Vigo , Ourense, Spain
| | - Alfonso Varela-Lopez
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy
| | - Jose L Quiles
- 4 Department of Physiology, Institute of Nutrition and Food Technology "Jose Mataix," Biomedical Research Centre, University of Granada , Granada, Spain
| | - Maurizio Battino
- 1 Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche-Sez. Biochimica , Facoltà di Medicina, Università Politecnica delle Marche , Ancona, Italy .,5 Centre for Nutrition and Health, Universidad Europea del Atlantico (UEA) , Santander, Spain
| |
Collapse
|
44
|
Huang Y, Xiong H, Chen K, Zhu X, Yin X, Liang Y, Luo W, Lei Q. [Inhibition of autophagy suppresses osteogenic differentiation of stem cells from apical papilla]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:106-112. [PMID: 30692075 DOI: 10.12122/j.issn.1673-4254.2019.01.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the effects of autophagy on osteogenic differentiation of stem cells from the apical papilla (SCAPs) in the presence of tumor necrosis factor-α (TNF-α) stimulation in vitro. METHODS SCAPs treated with TNF-α (0, 5, and 10 ng/mL) with or without 5 mmol/L 3-MA were examined for the expression of autophagy marker LC3-Ⅱ using Western blotting. The cells were transfected with GFP-LC3 plasmid and fluorescence microscopy was used for quantitative analysis of intracellular GFP-LC3; AO staining was used to detect the acidic vesicles in the cells. The cell viability was assessed with CCK-8 assays and the cell apoptosis rate was analyzed using flow cytometry. The cells treated with TNF-α or with TNF-α and 3-MA were cultured in osteogenic differentiation medium for 3 to 14 days, and real- time PCR was used to detect the mRNA expressions of osteogenesis-related genes (ALP, BSP, and OCN) for evaluating the cell differentiation. RESULTS TNF-α induced activation of autophagy in cultured SCAPs. Pharmacological inhibition of TNF-α-induced autophagy by 3-MA significantly decreased the cell viability and increased the apoptosis rate of SCAPs (P < 0.05). Compared with the cells treated with TNF-α alone, the cells treated with both TNF-α and 3-MA exhibited decreased expressions of the ALP and BSP mRNA on days 3, 7 and 14 during osteogenic induction (P < 0.05) and decreased expression of OCN mRNA on days 3 and 7 during the induction (P < 0.05). CONCLUSIONS Autophagy may play an important role during the osteogenic differentiation of SCAPs in the presence of TNF-α stimulation.
Collapse
Affiliation(s)
- Ying Huang
- Stomatology Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan 528308, China
| | - Huacui Xiong
- Stomatological Hospital, Southern Medical University, Guangzhou 510000, China.,Guangzhou Women and Children's Medical Center, Guangzhou 510623, China
| | - Ke Chen
- Stomatological Hospital, Southern Medical University, Guangzhou 510000, China.,Guangzhou Women and Children's Medical Center, Guangzhou 510623, China
| | - Xiaobin Zhu
- Stomatological Hospital, Southern Medical University, Guangzhou 510000, China
| | - Xiaoping Yin
- Affiliated Hospital of Guilin Medical University, Guilin 541000, China
| | - Yun Liang
- Stomatological Hospital, Southern Medical University, Guangzhou 510000, China.,Guangzhou Women and Children's Medical Center, Guangzhou 510623, China
| | - Wei Luo
- Guangzhou Women and Children's Medical Center, Guangzhou 510623, China
| | - Qiyin Lei
- Guangzhou Women and Children's Medical Center, Guangzhou 510623, China
| |
Collapse
|
45
|
Tao M, Shi Y, Tang L, Wang Y, Fang L, Jiang W, Lin T, Qiu A, Zhuang S, Liu N. Blockade of ERK1/2 by U0126 alleviates uric acid-induced EMT and tubular cell injury in rats with hyperuricemic nephropathy. Am J Physiol Renal Physiol 2019; 316:F660-F673. [PMID: 30648910 DOI: 10.1152/ajprenal.00480.2018] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Extracellular signal-regulated kinases 1 and 2 (ERK1/2) are serine/threonine kinases and function as regulators of cellular proliferation and differentiation. Recently, we demonstrated that inhibition of ERK1/2 alleviates the development and progression of hyperuricemia nephropathy (HN). However, its potential roles in uric acid-induced tubular epithelial-mesenchymal transition (EMT) and tubular epithelial cell injury are unknown. In this study, we showed that hyperuricemic injury induced EMT as characterized by downregulation of E-cadherin and upregulation of vimentin and Snail1 in a rat model of HN. This was coincident with epithelial cells arrested at the G2/M phase of cell cycle, activation of Notch1/Jagged-1 and Wnt/β-catenin signaling pathways, and upregulation of matrix metalloproteinase-2 (MMP-2) and MMP-9. Administration of U0126, a selective inhibitor of ERK1/2, blocked all these responses. U0126 was also effective in inhibiting renal tubular cell injury, as shown by decreased expression of lipocalin-2 and kidney injury molecule-1 and active forms of caspase-3. U0126 or ERK1/2 siRNA can inhibit tubular cell EMT and cell apoptosis as characterized with decreased expression of cleaved caspase-3. Moreover, ERK1/2 inhibition suppressed hyperuricemic injury-induced oxidative stress as indicated by decreased malondialdehyde and increased superoxide dismutase. Collectively, ERK1/2 inhibition-elicited renal protection is associated with inhibition of EMT through inactivation of multiple signaling pathways and matrix metalloproteinases, as well as attenuation of renal tubule injury by enhancing cellular resistance to oxidative stress.
Collapse
Affiliation(s)
- Min Tao
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine , Shanghai , China
| | - Yingfeng Shi
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine , Shanghai , China
| | - Lunxian Tang
- Emergency Department of Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine , Shanghai , China
| | - Yi Wang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine , Shanghai , China
| | - Lu Fang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine , Shanghai , China
| | - Wei Jiang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine , Shanghai , China
| | - Tao Lin
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine , Shanghai , China
| | - Andong Qiu
- School of Life Science and Technology, Advanced Institute of Translational Medicine, Tongji University , Shanghai , China
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine , Shanghai , China.,Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University , Providence, Rhode Island
| | - Na Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine , Shanghai , China
| |
Collapse
|
46
|
Escobar SJDM, Fong GM, Winnischofer SMB, Simone M, Munoz L, Dennis JM, Rocha MEM, Witting PK. Anti-proliferative and cytotoxic activities of the flavonoid isoliquiritigenin in the human neuroblastoma cell line SH-SY5Y. Chem Biol Interact 2018; 299:77-87. [PMID: 30502331 DOI: 10.1016/j.cbi.2018.11.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 11/26/2018] [Accepted: 11/27/2018] [Indexed: 12/15/2022]
Abstract
Neuroblastoma is a common childhood cancer with high mortality. We evaluated the capacity of the flavonoid, isoliquiritigenin (4,2',4'-trihydroxychalcone; ISL) to inhibit cellular proliferation and migration in the human neuroblastoma cell line SH-SY5Y. Incubation of cultured SH-SY5Y cells with 20-100 μM ISL decreased cell confluency (15-70%) after 24 h incubation, while 10-100 μM ISL (24 h) depleted intracellular ATP stores (15-90% vs vehicle-treated control) after 24 h incubation. ISL-mediated cell toxicity did not involve intracellular caspase 3/7 activation, externalization of phosphatidylserine on the cell membrane or stimulation of TNF and IL-1β release, all indicating that the flavonoid did not induce apoptosis. Pre-treatment of cells with necrostatin-1, a necroptosis inhibitor, significantly restored ATP levels (ATP levels increased 12-42%) in ISL-treated neuroblastoma cells indicative of enhanced viability. By contrast, RIP1 phosphorylation status remained unchanged in cells treated with ISL although the intracellular ratio of phosphorylated/total parental RIP1 increased after ISL treatment on SH-SY5Y cells indicating that ISL decreased levels of native RIP1. In addition, ISL treatment inhibited SH-SY5Y cell migration/proliferation in a scratch assay and arrested cell cycle transition by significantly decreasing the number of cells in G0/G1 phase and increasing populations by ~10% in S (primarily) and G2/M (lesser extent) phases. The intracellular ratio of phosphorylated/total ERK 1/2 and p38 remained unchanged after ISL treatment (up to 40 μM); ERK activation was only determined at ISL dose well above the experimental IC50 value as judged by ELISA analyses and this did not correlate with ISL cytotoxicity at lower dose <40 μM; Western blot assay confirmed the detection of phosphorylated (p-)ERK1/2 and (p-)p38 in ISL treated cells. Together the results suggest that ISL exerts anti-proliferative and cytotoxic activity on SHSY5Y cells through the loss of ATP, induction of cell cycle arrest, and cell death largely via a necroptotic mechanism in the absence of apoptotic activity.
Collapse
Affiliation(s)
- Stephane J de M Escobar
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, PR, Brazil; Redox Biology, Discipline of Pathology, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Genevieve M Fong
- Redox Biology, Discipline of Pathology, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Sheila M B Winnischofer
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Martin Simone
- Redox Biology, Discipline of Pathology, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Lenka Munoz
- Neuropharmacology Groups, Discipline of Pathology, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Joanne M Dennis
- Redox Biology, Discipline of Pathology, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Maria Eliane M Rocha
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Paul K Witting
- Redox Biology, Discipline of Pathology, The University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
47
|
Yushan R, Ying Y, Yujun T, Jingchun Y, Dongguang L, Lihong P, Pingping W, Lili Z, Fanhui Z, Zhong L, Guimin Z, Jie L. Isoliquiritigenin inhibits mouse S180 tumors with a new mechanism that regulates autophagy by GSK-3β/TNF-α pathway. Eur J Pharmacol 2018; 838:11-22. [DOI: 10.1016/j.ejphar.2018.08.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 08/21/2018] [Accepted: 08/24/2018] [Indexed: 11/26/2022]
|
48
|
Ge Y, Huang M, Yao YM. Autophagy and proinflammatory cytokines: Interactions and clinical implications. Cytokine Growth Factor Rev 2018; 43:38-46. [PMID: 30031632 DOI: 10.1016/j.cytogfr.2018.07.001] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 07/10/2018] [Accepted: 07/11/2018] [Indexed: 02/07/2023]
Abstract
Autophagy is a ubiquitous cellular process that regulates cell growth, survival, development and death. Its process is closely associated with diverse conditions, such as liver diseases, neurodegenerative diseases, myopathy, heart diseases, cancer, immunization, and inflammatory diseases. Thus, understanding the modulation of autophagy may provide novel insight into potential therapeutic targets. Autophagy is closely intertwined with inflammatory and immune responses, and cytokines may help mediate this interaction. Autophagy has been shown to regulate, and be regulated by, a wide range of proinflammatory cytokines. This review aims to summarize recent progress in elucidating the interplay between autophagy and proinflammatory cytokines, including IFN-γ, TNF-α, IL-17, and cytokines of the IL-1 family (e.g., IL-1α, IL-1β, IL-33, and IL-36).
Collapse
Affiliation(s)
- Yun Ge
- Department of General Intensive Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Man Huang
- Department of General Intensive Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yong-Ming Yao
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, No.51 Fu-Cheng Road, Beijing 100048, China.
| |
Collapse
|
49
|
Kaneko H, Kobayashi M, Mizunoe Y, Yoshida M, Yasukawa H, Hoshino S, Itagawa R, Furuichi T, Okita N, Sudo Y, Imae M, Higami Y. Taurine is an amino acid with the ability to activate autophagy in adipocytes. Amino Acids 2018. [DOI: 10.1007/s00726-018-2550-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
50
|
Isoliquiritigenin induces apoptosis and autophagy and inhibits endometrial cancer growth in mice. Oncotarget 2018; 7:73432-73447. [PMID: 27708238 PMCID: PMC5341989 DOI: 10.18632/oncotarget.12369] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 09/25/2016] [Indexed: 12/17/2022] Open
Abstract
Endometrial cancer is the most common cancer in women, typically with onset after menopause. Isoliquiritigenin (ISL), a licorice flavonoid, was previously shown to have anti-oxidant, anti-inflammatory, and tumor suppression effects. In this study, we investigated the anti-tumor effect of ISL on human endometrial cancer both in vitro and in vivo. We used telomerase-immortalized human endometrial stromal cells (T-HESCs) and human endometrial cancer cell lines (Ishikawa, HEC-1A, and RL95-2 cells) as targets. The effects of ISL on cell proliferation, cell cycle regulation, and apoptosis or autophagy-related protein expression were examined. In addition, we conducted in vivo experiments to confirm the inhibitory effects of ISL on cancer cells. ISL significantly inhibited the viability of cancer cells in a dose- and time-dependent manner but with little toxicity on normal cells. In addition, flow cytometry analysis indicated that ISL induced sub-G1 or G2/M phase arrest. ISL treatment activated the extracellular signal regulated kinase signaling pathway to enhance the protein expression of caspase-7/LC3BII associated with apoptosis/autophagy. Furthermore, ISL suppressed xenograft tumor growth in vivo. Taken together, these findings suggest that ISL may induce apoptosis, autophagy, and cell growth inhibition, indicating its potential as a therapeutic agent for human endometrial cancer.
Collapse
|