1
|
Chen P, Zhang X, Fang Q, Zhao Z, Lin C, Zhou Y, Liu F, Zhu C, Wu A. Betulinic acid induces apoptosis of HeLa cells via ROS-dependent ER stress and autophagy in vitro and in vivo. J Nat Med 2024; 78:677-692. [PMID: 38403724 DOI: 10.1007/s11418-024-01782-6] [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: 09/19/2023] [Accepted: 01/16/2024] [Indexed: 02/27/2024]
Abstract
Betulinic acid (BA), a naturally occurring lupane-type triterpenoid, possesses a wide range of potential activities against different types of cancer. However, the molecular mechanisms involved in anti-cervical cancer about BA were rarely investigated. Herein, the role of BA in cervical cancer suppression by ROS-mediated endoplasmic reticulum stress (ERS) and autophagy was deeply discussed. The findings revealed that BA activated Keap1/Nrf2 pathway and triggered mitochondria-dependent apoptosis due to ROS production. Furthermore, BA increased the intracellular Ca2+ levels, inhibited the expression of Beclin1 and promoted the expression of GRP78, LC3-II, and p62 associated with ERS and autophagy. Besides, BA initiated the formation of autophagosomes and inhibited autophagic flux by the co-administration of BA with 3-methyladenine (3-MA) and chloroquine (CQ), respectively. The in vivo experiment manifested that hydroxychloroquine (HCQ) enhanced the apoptosis induced by BA. For the first time, we demonstrated that BA could initiate early autophagy, inhibit autophagy flux, and induce protective autophagy in HeLa cells. Thus, BA could be a potential chemotherapy drug for cervical cancer, and inhibition of autophagy could enhance the anti-tumor effect of BA. However, the interactions of signaling factors between ERS-mediated and autophagy-mediated apoptosis deserve further attention.
Collapse
Affiliation(s)
- Ping Chen
- School of Pharmaceutical Sciences, GuangZhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Xueer Zhang
- School of Pharmaceutical Sciences, GuangZhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Qiaomiao Fang
- School of Pharmaceutical Sciences, GuangZhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Zhongxiang Zhao
- School of Pharmaceutical Sciences, GuangZhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Chaozhan Lin
- School of Pharmaceutical Sciences, GuangZhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Yuan Zhou
- School of Pharmaceutical Sciences, GuangZhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Fangle Liu
- School of Pharmaceutical Sciences, GuangZhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Chenchen Zhu
- School of Pharmaceutical Sciences, GuangZhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Aizhi Wu
- School of Pharmaceutical Sciences, GuangZhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China.
| |
Collapse
|
2
|
Zhang Q, Liu G, Liu R, Liu J, Zeng X, Ren D, Yan X, Yuan X. Dual role of endoplasmic reticulum stress-ATF-6 activation in autophagy and apoptosis induced by cyclic stretch in myoblast. Apoptosis 2023; 28:796-809. [PMID: 36881290 DOI: 10.1007/s10495-023-01825-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2023] [Indexed: 03/08/2023]
Abstract
OBJECTIVE Mandibular growth that is induced by functional appliances is closely associated with skeletal and neuromuscular adaptation. Accumulating evidence has proved that apoptosis and autophagy have a vital role in adaptation process. However, little is known about the underlying mechanisms. This study sought to determine whether ATF-6 is involved in stretch-induced apoptosis and autophagy in myoblast. The study also sought to uncover the potential molecular mechanism. MATERIALS AND METHODS Apoptosis was assessed by TUNEL and Annexin V and PI staining. Autophagy was detected by transmission electron microscopy (TEM) analysis and immunofluorescent staining for autophagy-related protein light chain 3 (LC3). Real time-PCR and western blot were performed to evaluate the expression level of mRNA and proteins that were associated with endoplasmic reticulum stress (ERS), autophagy and apoptosis. RESULTS Cyclic stretch significantly decreased the cell viability and induced apoptosis and autophagy of myoblasts time-dependently. Stretching stimuli activated ATF-6 pathway and induced ERS-mediated apoptosis. Moreover, using 4-PBA significantly inhibited ERS-related apoptosis, as well as partially decreasing autophagy. In addition, inhibition of autophagy by 3-MA enhanced apoptosis by affecting the expression of CHOP and Bcl-2. However, it had no obvious effects on ERS-related proteins of GRP78 and ATF-6. More importantly, knockdown ATF-6 effectively weakened apoptosis and autophagy. It did so by regulating the expression of Bcl-2, Beclin1 and CHOP, but not cleaved Caspase-12, LC3II and p62 in stretched myoblast. CONCLUSION ATF-6 pathway was activated in myoblast by mechanical stretch. ATF-6 may regulate the process of stretch-induced myoblast apoptosis and autophagy via CHOP, Bcl-2 and Beclin1 signaling.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology, Qingdao University, Qingdao, China
| | - Guirong Liu
- Department of Stomatology, Qingdao Municipal Hospital, Qingdao, China
| | - Ran Liu
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jun Liu
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology, Qingdao University, Qingdao, China
| | - Xuemin Zeng
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology, Qingdao University, Qingdao, China
| | - Dapeng Ren
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology, Qingdao University, Qingdao, China
| | - Xiao Yan
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China. .,School of Stomatology, Qingdao University, Qingdao, China.
| | - Xiao Yuan
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China. .,School of Stomatology, Qingdao University, Qingdao, China.
| |
Collapse
|
3
|
Nokhostin F, Azadehrah M, Azadehrah M. The multifaced role and therapeutic regulation of autophagy in ovarian cancer. Clin Transl Oncol 2022; 25:1207-1217. [PMID: 36534371 DOI: 10.1007/s12094-022-03045-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
Ovarian cancer (OC) is one of the tumors that occurs most frequently in women. Autophagy is involved in cell homeostasis, biomolecule recycling, and survival, making it a potential target for anti-tumor drugs. It is worth noting that growing evidence reveals a close link between autophagy and OC. In the context of OC, autophagy demonstrates activity as both a tumor suppressor and a tumor promoter, depending on the context. Autophagy's exact function in OC is greatly reliant on the tumor microenvironment (TME) and other conditions, such as hypoxia, nutritional deficiency, chemotherapy, and so on. However, what can be concluded from different studies is that autophagy-related signaling pathways, especially PI3K/AKT/mTOR axis, increase in advanced stages and malignant phenotype of the disease reduces autophagy and ultimately leads to tumor progression. This study sought to present a thorough understanding of the role of autophagy-related signaling pathways in OC and existing therapies targeting these signaling pathways.
Collapse
Affiliation(s)
- Fahimeh Nokhostin
- Department of Obstetrics and Gynecology, Faculty of Medicine, Shahid Sadughi University of Medical Sciences, Yazd, Iran
| | - Mahboobeh Azadehrah
- Cancer Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Malihe Azadehrah
- Cancer Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
4
|
Cong L, Bai Y, Guo Z. The crosstalk among autophagy, apoptosis, and pyroptosis in cardiovascular disease. Front Cardiovasc Med 2022; 9:997469. [PMID: 36386383 PMCID: PMC9650365 DOI: 10.3389/fcvm.2022.997469] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/10/2022] [Indexed: 08/02/2023] Open
Abstract
In recent years, the mechanism of cell death has become a hotspot in research on the pathogenesis and treatment of cardiovascular disease (CVD). Different cell death modes, including autophagy, apoptosis, and pyroptosis, are mosaic with each other and collaboratively regulate the process of CVD. This review summarizes the interaction and crosstalk of key pathways or proteins which play a critical role in the entire process of CVD and explores the specific mechanisms. Furthermore, this paper assesses the interrelationships among these three cell deaths and reviews how they regulate the pathogenesis of CVD. By understanding how these three cell death modes go together we can learn about the pathogenesis of CVD, which will enable us to identify new targets for preventing, controlling, and treating CVD. It will not only reduce mortality but also improve the quality of life.
Collapse
Affiliation(s)
- Lin Cong
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Department of Cardiac Surgery, Chest Hospital, Tianjin University, Tianjin, China
| | - Yunpeng Bai
- Department of Cardiac Surgery, Chest Hospital, Tianjin University, Tianjin, China
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
| | - Zhigang Guo
- Department of Cardiac Surgery, Chest Hospital, Tianjin University, Tianjin, China
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
| |
Collapse
|
5
|
Over Fifty Years of Life, Death, and Cannibalism: A Historical Recollection of Apoptosis and Autophagy. Int J Mol Sci 2021; 22:ijms222212466. [PMID: 34830349 PMCID: PMC8618802 DOI: 10.3390/ijms222212466] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 01/18/2023] Open
Abstract
Research in biomedical sciences has changed dramatically over the past fifty years. There is no doubt that the discovery of apoptosis and autophagy as two highly synchronized and regulated mechanisms in cellular homeostasis are among the most important discoveries in these decades. Along with the advancement in molecular biology, identifying the genetic players in apoptosis and autophagy has shed light on our understanding of their function in physiological and pathological conditions. In this review, we first describe the history of key discoveries in apoptosis with a molecular insight and continue with apoptosis pathways and their regulation. We touch upon the role of apoptosis in human health and its malfunction in several diseases. We discuss the path to the morphological and molecular discovery of autophagy. Moreover, we dive deep into the precise regulation of autophagy and recent findings from basic research to clinical applications of autophagy modulation in human health and illnesses and the available therapies for many diseases caused by impaired autophagy. We conclude with the exciting crosstalk between apoptosis and autophagy, from the early discoveries to recent findings.
Collapse
|
6
|
Li Y, Gao S, Du X, Ji J, Xi Y, Zhai G. Advances in autophagy as a target in the treatment of tumours. J Drug Target 2021; 30:166-187. [PMID: 34319838 DOI: 10.1080/1061186x.2021.1961792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Autophagy is a multi-step lysosomal degradation process, which regulates energy and material metabolism and has been used to maintain homeostasis. Autophagy has been shown to be involved in the regulation of health and disease. But at present, there is no consensus on the relationship between autophagy and tumour, and we consider that it plays a dual role in the occurrence and development of tumour. That is to say, under certain conditions, it can inhibit the occurrence of tumour, but it can also promote the process of tumour. Therefore, autophagy could be used as a target for tumour treatment. The regulation of autophagy plays a synergistic role in the radiotherapy, chemotherapy, phototherapy and immunotherapy of tumour, and nano drug delivery system provides a promising strategy for improving the efficacy of autophagy regulation. This review summarised the progress in the regulatory pathways and factors of autophagy as well as nanoformulations as carriers for the delivery of autophagy modulators.
Collapse
Affiliation(s)
- Yingying Li
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Shan Gao
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Xiyou Du
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Yanwei Xi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| |
Collapse
|
7
|
Li C, Li Z, Song L, Meng L, Xu G, Zhang H, Hu J, Li F, Liu C. GEFT Inhibits Autophagy and Apoptosis in Rhabdomyosarcoma via Activation of the Rac1/Cdc42-mTOR Signaling Pathway. Front Oncol 2021; 11:656608. [PMID: 34221974 PMCID: PMC8252888 DOI: 10.3389/fonc.2021.656608] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/20/2021] [Indexed: 12/12/2022] Open
Abstract
Autophagy and apoptosis are dynamic processes that determine the fate of cells, and regulating these processes can treat cancer. GEFT is highly expressed in rhabdomyosarcoma (RMS), which accelerates the tumorigenicity and metastasis of RMS by activating Rac1/Cdc42 signaling, but the regulatory mechanisms of autophagy and apoptosis are unclear. In our study, we found that the RMS tissues had high Rac1, Cdc42, mTOR, and Bcl-2 expression levels and low Beclin1, LC3, and Bax expression levels compared with the normal striated muscle tissues (P < 0.05). In addition, multivariate analysis has proven that Rac1 is an independent prognostic factor (P < 0.05), and the high expression level of the Beclin1 protein was closely associated with the tumor diameter of the RMS patients (P = 0.044), whereas the high expression level of the LC3 protein was associated with the clinical stage of the RMS patients (P = 0.027). Furthermore, GEFT overexpression could inhibit autophagy and apoptosis in RMS. A Rac1/Cdc42 inhibitor was added, and the inhibition of autophagy and apoptosis decreased. Rac1 and Cdc42 could regulate mTOR to inhibit autophagy and apoptosis in RMS. Overall, these studies demonstrated that the GEFT–Rac1/Cdc42–mTOR pathway can inhibit autophagy and apoptosis in RMS and provide evidence for innovative treatments.
Collapse
Affiliation(s)
- Chunsen Li
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, China
| | - Zhenzhen Li
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, China
| | - Lingxie Song
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, China.,Department of Pathology and Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Lian Meng
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, China
| | - Guixuan Xu
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, China
| | - Haijun Zhang
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, China
| | - Jianming Hu
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, China
| | - Feng Li
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, China.,Department of Pathology and Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Chunxia Liu
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, China.,Department of Pathology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
8
|
Shen L, Xia M, Zhang Y, Luo H, Dong D, Sun L. Mitochondrial integration and ovarian cancer chemotherapy resistance. Exp Cell Res 2021; 401:112549. [PMID: 33640393 DOI: 10.1016/j.yexcr.2021.112549] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/21/2021] [Accepted: 02/23/2021] [Indexed: 10/22/2022]
Abstract
Ovarian cancer has been nicknamed the "silent killer". Most patients with ovarian cancer are diagnosed at an advanced stage of the disease for the first time because of its insignificant early clinical symptoms. In addition to the difficulty of early screening and delay in diagnosis, the high recurrence rate and relapsed refractory status of patients with ovarian cancer are also important factors for their high mortality. Patients with recurrent ovarian cancer often use neoadjuvant chemotherapy followed by surgery as the first choice. However, this is often accompanied by chemotherapy resistance, leading to treatment failure and a mortality rate of more than 90%. In the past, it was believed that the anti-tumor effect of chemotherapeutics represented by cisplatin was entirely attributable to its irreversible damage to DNA, but current research has found that it can inhibit cell growth and cytotoxicity via nuclear and cytoplasmic coordinated integration. As an important hub and integration platform for intracellular signal communication, mitochondria are responsible for multiple key factors during tumor occurrence and development, such as metabolic reprogramming, acquisition of metastatic ability, and chemotherapy drug response. The role of mitochondria in ovarian cancer chemotherapy resistance is becoming increasingly recognized. In this review, we discuss the cellular interactive regulatory network surrounding mitochondria, elucidate the mechanisms of tumor cell survival under chemotherapy, and discuss potential means of interfering with mitochondrial function as a novel anti-cancer therapy.
Collapse
Affiliation(s)
- Luyan Shen
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Meihui Xia
- Department of Obstetrics, The First Bethune Hospital of Jilin University, Changchun, Jilin, China
| | - Yu Zhang
- Laboratory Teaching Center of Basic Medicine, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Haoge Luo
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Delu Dong
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China.
| | - Liankun Sun
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China.
| |
Collapse
|
9
|
Tang Z, Chen H, Zhong D, Wei W, Liu L, Duan Q, Han B, Li G. TRIB3 facilitates glioblastoma progression via restraining autophagy. Aging (Albany NY) 2020; 12:25020-25034. [PMID: 33203798 PMCID: PMC7803516 DOI: 10.18632/aging.103969] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/21/2020] [Indexed: 12/21/2022]
Abstract
The pseudokinase Tribble 3 (TRIB3) is known as a regulator in cellular responses to a variety of stresses, such as glucose insufficiency and endoplasmic reticulum (ER) stress. TRIB3 is upregulated in various cancer tissues and is closely connected to the poor prognosis of patients. However, the underlying regulation and function of TRIB3 in glioblastoma (GBM) is still largely unknown. In this study, the upregulation of TRIB3 was confirmed both in primary specimens from GBM patients and in vitro with GBM cell lines. Overexpression of specific TRIB3 transcripts promoted cell growth and migration in vitro, while knockdown of TRIB3 expression exerted a repressive effect on these cellular processes. The growth-promoting effect of TRIB3 was also demonstrated in a xenograft mouse model. Mechanistic studies further revealed that TRIB3 was able to suppress autophagic flux and that this suppression was responsible for TRIB3 silencing-induced proliferation and migration of GBM cells. These findings indicate that the suppression of autophagic flux by TRIB3 drives the invasion and proliferation of GBM cells, thus suggesting that TRIB3 is a potential novel therapeutic target for the treatment of glioma.
Collapse
Affiliation(s)
- Zhanbin Tang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Hongping Chen
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Di Zhong
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Wan Wei
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Lili Liu
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Qiong Duan
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Baichao Han
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Guozhong Li
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
10
|
Shi Y, He R, Yang Y, He Y, Zhan L, Wei B. Potential relationship between Sirt3 and autophagy in ovarian cancer. Oncol Lett 2020; 20:162. [PMID: 32934730 PMCID: PMC7471650 DOI: 10.3892/ol.2020.12023] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 07/30/2020] [Indexed: 02/06/2023] Open
Abstract
Sirtuin 3 (Sirt3) is an important member of the sirtuin protein family. It is a deacetylase that was previously reported to modulate the level of reactive oxygen species (ROS) production and limit the extent of oxidative damage in cellular components. As an important member of the class III type of histone deacetylases, Sirt3 has also been documented to mediate nuclear gene expression, metabolic control, neuroprotection, cell cycle and proliferation. In ovarian cancer (OC), Sirt3 has been reported to regulate cellular metabolism, apoptosis and autophagy. Sirt3 can regulate autophagy through a variety of different molecular signaling pathways, including the p62, 5'AMP-activated protein kinase and mitochondrial ROS-superoxide dismutase pathways. However, autophagy downstream of Sirt3 and its association with OC remains poorly understood. In the present review, the known characteristics of Sirt3 and autophagy were outlined, and their potential functional roles were discussed. Following a comprehensive analysis of the current literature, Sirt3 and autophagy may either serve positive or negative roles in the regulation of OC. Therefore, it is important to identify the appropriate expression level of Sirt3 to control the activation of autophagy in OC cells. This strategy may prove to be a novel therapeutic method to reduce the mortality of patients with OC. Finally, potential research directions into the association between Sirt3 and other signaling pathways were provided.
Collapse
Affiliation(s)
- Yuchuan Shi
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Runhua He
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Yu Yang
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Yu He
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Lei Zhan
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China.,Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Bing Wei
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| |
Collapse
|
11
|
Wang P, Zhao ZQ, Guo SB, Yang TY, Chang ZQ, Li DH, Zhao W, Wang YX, Sun C, Wang Y, Feng W. Roles of microRNA-22 in Suppressing Proliferation and Promoting Sensitivity of Osteosarcoma Cells via Metadherin-mediated Autophagy. Orthop Surg 2019; 11:285-293. [PMID: 30932352 PMCID: PMC6594522 DOI: 10.1111/os.12442] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/21/2019] [Accepted: 02/24/2019] [Indexed: 12/14/2022] Open
Abstract
Objective To analyze the effect of microRNA‐22 on autophagy and proliferation and to investigate the underlying molecular mechanism of osteosarcoma cell chemotherapy sensitivity. Methods MG‐63 cells were divided into four groups, including a control group, a negative control (NC) group, a cisplatin group, and a cisplatin + miR‐22 group. Proliferation of MG‐63 cells that had been treated with cisplatin and transfected with miR‐22 mimics was determined using MTT assay and colony formation assay. We assessed the degree of autophagy using flow cytometry through cellular staining of the autophagy lysosomal marker monodansylcadaverine (MDC). The effect of microRNA‐22 on autophagy was observed along with the expression levels of Beclin1, LC3, metadherin (MTDH) and ATG5 by western blot and quantitative reverse transcription polymerase chain reaction (qRT‐PCR). Luciferase reporter assay revealed the targeted binding site between miR‐22 and the 3′‐untranslated region (3′‐UTR) of MTDH mRNA. Western blot and qRT‐PCR were used to explore the level of MTDH in the control group, the NC group, the cisplatin group, and the miR‐22 group for 6, 12, and 24 h. Results In the in vitro study, the MTT results indicated that the MG‐63 cells with overexpression of miR‐22 exhibited a significant decline in the proliferation capacity compared with the control group (0.513 ± 0.001, P < 0.0005). Similar to the MTT results, MG‐63 cells that were transfected with miR‐22 mimic (101.0 ± 10.58) formed fewer colonies compared with the cisplatin group (129.7 ± 4.163). MDC staining revealed that miR‐22‐overexpressing osteosarcoma (OS) cells treated with cisplatin showed a significant decrease in the expression of autophagy (7.747 ± 0.117, P < 0.0001). Our data revealed that miR‐22 could regulate not only autophagy but also proliferation in the chemosensitivity of osteosarcoma cells. We found that miR‐22 sensitized osteosarcoma cells to cisplatin treatment by regulating autophagy‐related genes. In addition, Luciferase Reporter Assay revealed that miR‐22 negatively regulated autophagy through direct targeting of MTDH. We performed western blot analysis to detect the MTDH expression level. The results revealed that the overexpression of miR‐22 obviously decreased the expression of MTDH (1.081 ± 0.023, P < 0.001). Conclusion Inhibition of miR‐22 ameliorated the anticancer drug‐induced cell proliferation decrease in osteosarcoma cells. MTDH was identified as the miR‐22 target in OS cells and MTDH‐triggered autophagy played a key function in the miR‐22‐associated chemotherapy sensitivity.
Collapse
Affiliation(s)
- Peng Wang
- Orthopedics Department, Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Zhen-Qun Zhao
- Orthopedics Department, Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Shi-Bing Guo
- Orthopedics Department, Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Tie-Yi Yang
- Orthopedics Department, Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Zhi-Qiang Chang
- Orthopedics Department, Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Dai-He Li
- Orthopedics Department, Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Wei Zhao
- Orthopedics Department, Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yu-Xin Wang
- Orthopedics Department, Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Chao Sun
- Orthopedics Department, Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yong Wang
- Orthopedics Department, Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Wei Feng
- Orthopedics Department, Inner Mongolia Institute of Orthopaedics, Hohhot, China
| |
Collapse
|
12
|
Zhou L, Kong Q, Zhang Y, Yang W, Yan S, Li M, Wang Y, Zhou Y, Yu H, Han L. Glucose deprivation promotes apoptotic response to S1 by enhancing autophagy in human cervical cancer cells. Cancer Manag Res 2018; 10:6195-6204. [PMID: 30538566 PMCID: PMC6260140 DOI: 10.2147/cmar.s184180] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background S1 is a novel BH3 mimetic that can induce death in some types of cancer cells, such as melanoma B16, ovarian cancer SKOV3, and U251 glioma cells. S1 inhibits Bcl-2 and Mcl-1 expression and induces cancer cell apoptosis. Cancer cell survival is highly dependent on glucose. Here, we observed the effect of glucose deprivation on the apoptotic response to S1 in human cervical cancer (HeLa) cells. Materials and methods Earle’s balanced salt solution (EBSS) was used to simulate glucose deprivation. MTT assay was used to analyze the cell survival rate, and Hoechst 33342 dye was used to detect the apoptosis in HeLa cells. Western blotting was used to detect the expression of ER stress and autophagy relative proteins. In addition, lysosomes were observed with Lyso-Tracker staining by confocal microscopy. Results S1 decreased cell distribution density and survival rate, and MTT assay showed that EBSS enhanced the inhibitory effects of S1 on HeLa cell growth. Hoechst 33342 dye showed that S1 led to pyknosis, fragmentation, and strong staining in HeLa cell nuclei, and EBSS enhanced these effects. Western blotting indicated that EBSS enhanced the expression of apoptosis-related proteins (cytochrome C, caspase-3, and poly[ADP-ribose] polymerase 1) induced by S1 in HeLa cells. S1 decreased p62 expression and increased the autophagosome-associated protein LC3-II expression, which indicated that S1 induced autophagy in HeLa cells. EBSS enhanced S1-induced autophagy in HeLa cells. Furthermore, autophagy inhibitor chloroquine enhanced S1-induced apoptosis in HeLa cells. Conclusion These results indicate that EBSS exacerbates S1-induced autophagy and severe autophagy induced by EBSS and S1 could lead to apoptosis in HeLa cells. The results also suggest that EBSS enhances the sensitivity of HeLa cells to S1-induced apoptosis and that autophagy plays an important role in this process.
Collapse
Affiliation(s)
- Li Zhou
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130041, China, .,Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun 130021, China
| | - Qinghuan Kong
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Jilin University, Changchun 130021, China,
| | - Yunhan Zhang
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Jilin University, Changchun 130021, China,
| | - Wei Yang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun 130021, China
| | - Shan Yan
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Jilin University, Changchun 130021, China,
| | - Meihui Li
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Jilin University, Changchun 130021, China,
| | - Yidan Wang
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Jilin University, Changchun 130021, China,
| | - Yanjie Zhou
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Jilin University, Changchun 130021, China,
| | - Huimei Yu
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Jilin University, Changchun 130021, China,
| | - Liying Han
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130041, China,
| |
Collapse
|
13
|
Sophia J, Kowshik J, Dwivedi A, Bhutia SK, Manavathi B, Mishra R, Nagini S. Nimbolide, a neem limonoid inhibits cytoprotective autophagy to activate apoptosis via modulation of the PI3K/Akt/GSK-3β signalling pathway in oral cancer. Cell Death Dis 2018; 9:1087. [PMID: 30352996 PMCID: PMC6199248 DOI: 10.1038/s41419-018-1126-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/24/2018] [Accepted: 10/04/2018] [Indexed: 02/08/2023]
Abstract
Of late, nimbolide, a limonoid from the neem tree (Azadirachta indica) has gained increasing research attention owing to its potent antiproliferative and apoptosis-inducing effects. The present study was designed to investigate the effect of nimbolide on autophagy and the time point at which the phosphorylation status of GSK-3β and PI3K dictate the choice between autophagy and apoptosis in SCC131 and SCC4 oral cancer cells. Additionally, we analysed changes in the expression of proteins involved in autophagy and apoptosis after therapeutic intervention with nimbolide in a hamster model of oral oncogenesis. Furthermore, we also demonstrate changes in the expression of key genes involved in apoptosis and autophagy during the stepwise evolution of hamster and human OSCCs. Nimbolide-induced stereotypical changes in oral cancer cells characteristic of both apoptosis and autophagy. Time-course experiments revealed that nimbolide induces autophagy as an early event and then switches over to apoptosis. Nimbolide negatively regulates PI3K/Akt signalling with consequent increase in p-GSK-3βTyr216, the active form of GSK-3β that inhibits autophagy. Downregulation of HOTAIR, a competing endogenous RNA that sponges miR-126 may be a major contributor to the inactivation of PI3K/Akt/GSK3 signalling by nimbolide. Analysis of key markers of apoptosis and autophagy as well as p-AktSer473 during sequential progression of hamster and human OSCC revealed a gradual evolution to a pro-autophagic and antiapoptotic phenotype that could confer a survival advantage to tumors. In summary, the results of the present study provide insights into the molecular mechanisms by which nimbolide augments apoptosis by overcoming the shielding effects of cytoprotective autophagy through modulation of the phosphorylation status of Akt and GSK-3β as well as the ncRNAs miR-126 and HOTAIR. Development of phytochemicals such as nimbolide that target the complex interaction between proteins and ncRNAs that regulate the autophagy/apoptosis flux is of paramount importance in cancer prevention and therapeutics.
Collapse
Affiliation(s)
- Josephraj Sophia
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, 608 002, Tamil Nadu, India
| | - Jaganathan Kowshik
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, 608 002, Tamil Nadu, India
| | - Anju Dwivedi
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Sujit K Bhutia
- Department of Life Science, National Institute of Technology Rourkela, Rourkela, 769008, Odisha, India
| | - Bramanandam Manavathi
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Rajakishore Mishra
- Centre for Life Sciences, School of Natural Sciences, Central University of Jharkhand, Ranchi, 835205, Jharkhand, India
| | - Siddavaram Nagini
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, 608 002, Tamil Nadu, India.
| |
Collapse
|
14
|
Zhan L, Zhang Y, Wang W, Song E, Fan Y, Li J, Wei B. Autophagy as an emerging therapy target for ovarian carcinoma. Oncotarget 2018; 7:83476-83487. [PMID: 27825125 PMCID: PMC5347782 DOI: 10.18632/oncotarget.13080] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/21/2016] [Indexed: 12/11/2022] Open
Abstract
Autophagy is a conserved cellular self-digestion pathway for maintenance of homeostasis under basal and stressed conditions. Autophagy plays pivotal roles in the pathogenesis of many diseases, such as aging-related diseases, autoimmune diseases, cardiovascular diseases, and cancers. Of special note is that accumulating data suggest an intimate relationship between autophagy and ovarian carcinoma. Autophagy is well identified to act as either as a tumor-suppressor or as a tumor-promoter in ovarian carcinoma. The exact function of autophagy in ovarian carcinoma is highly dependent on the circumstances of cancer including hypoxic, nutrient-deficient, chemotherapy and so on. However, the mechanism underlying autophagy associated with ovarian carcinoma remains elusive, the precise role of autophagy in ovarian carcinoma also remains undetermined. In this review, we tried to sum up and discuss recent research achievements of autophagy in ovarian cancer. Moreover, waves of novel therapies ways for ovarian carcinoma based on the functions of autophagy were collected.
Collapse
Affiliation(s)
- Lei Zhan
- Department of gynecology and obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yu Zhang
- Department of gynecology and obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Wenyan Wang
- Department of gynecology and obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Enxue Song
- Department of gynecology and obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yijun Fan
- Department of gynecology and obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Jun Li
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China
| | - Bing Wei
- Department of gynecology and obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| |
Collapse
|
15
|
Yu G, Li N, Zhao Y, Wang W, Feng XL. Salidroside induces apoptosis in human ovarian cancer SKOV3 and A2780 cells through the p53 signaling pathway. Oncol Lett 2018; 15:6513-6518. [PMID: 29616120 DOI: 10.3892/ol.2018.8090] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 11/16/2017] [Indexed: 12/15/2022] Open
Abstract
Salidroside is one of the most potent compounds extracted from the plant Rhodiola rosea, and its cardiovascular protective effects have been studied extensively. However, the role of salidroside in human ovarian carcinoma remains unknown. The aim of the current study was to investigate the effects of salidroside on the proliferation and apoptosis of SKOV3 and A2780 cells using MTT assay and acridine orange/ethidium bromide staining. Salidroside activated caspase-3 and upregulated the levels of apoptosis-inducing factor, Bcl-2-associated X and Bcl-2-associated death promoter (Bad) proteins. Furthermore, salidroside downregulated the levels of Bcl-2, p-Bad and X-linked inhibitor of apoptosis proteins. Salidroside activated the caspase-dependent pathway in SKOV3 and A2780 cells, upregulating p53, p21Cip1/Waf1 and p16INK4a. These results suggest that the p53/p21Cip1/Waf1/p16INK4a pathway may serve a key function in salidroside-mediated effects on SKOV3 and A2780 cells. The current findings indicate that salidroside may be a promising novel drug candidate for ovarian cancer therapy.
Collapse
Affiliation(s)
- Ge Yu
- Department of Gynecology of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150001, P.R. China
| | - Na Li
- Department of Gynecology of Traditional Chinese Medicine, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150001, P.R. China
| | - Yan Zhao
- Department of Gynecology of Traditional Chinese Medicine, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150001, P.R. China
| | - Wei Wang
- Department of Gynecology of Traditional Chinese Medicine, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150001, P.R. China
| | - Xiao-Ling Feng
- Department of Gynecology of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150001, P.R. China.,Department of Gynecology of Traditional Chinese Medicine, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
16
|
Lu Z, Chen C, Wu Z, Miao Y, Muhammad I, Ding L, Tian E, Hu W, Ni H, Li R, Wang B, Li J. A Dual Role of P53 in Regulating Colistin-Induced Autophagy in PC-12 Cells. Front Pharmacol 2017; 8:768. [PMID: 29163157 PMCID: PMC5664992 DOI: 10.3389/fphar.2017.00768] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/11/2017] [Indexed: 12/15/2022] Open
Abstract
This study aimed to investigate the mechanism of p53 in regulating colistin-induced autophagy in PC-12 cells. Importantly, cells were treated with 125 μg/ml colistin for 12 and 24 h after transfection with p53 siRNA or recombinant plasmid. The hallmarks of autophagy and apoptosis were examined by real-time PCR and western blot, fluorescence/immunofluorescence microscopy, and electron microscopy. The results showed that silencing of p53 leads to down-regulation of Atg5 and beclin1 for 12 h while up-regulation at 24 h and up-regulation of p62 noted. The ratio of LC3-II/I and autophagic vacuoles were significantly increased at 24 h, but autophagy flux was blocked. The cleavage of caspase3 and PARP (poly ADP-ribose polymerase) were enhanced, while PC-12-sip53 cells exposed to 3-MA showed down-regulation of apoptosis. By contrast, the expression of autophagy-related genes and protein reduced in p53 overexpressing cells following a time dependent manner. Meanwhile, there was an increase in the expression of activated caspase3 and PARP, condensed and fragmented nuclei were evident. Conclusively, the data supported that silencing of p53 promotes impaired autophagy, which acts as a pro-apoptotic induction factor in PC-12 cells treated with colistin for 24 h, and overexpression of p53 inhibits autophagy and accelerates apoptosis. Hence, it has been suggested that p53 could not act as a neuro-protective target in colistin-induced neurotoxicity.
Collapse
Affiliation(s)
- Ziyin Lu
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Department of Animal Production, College of Life Engineering, Shenyang Institute of Technology, Fushun, China
| | - Chunli Chen
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zhiyong Wu
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yusong Miao
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Ishfaq Muhammad
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Liangjun Ding
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Erjie Tian
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Wanjun Hu
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Huilin Ni
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Rui Li
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Bo Wang
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Jichang Li
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| |
Collapse
|
17
|
(18-Crown-6)potassium(I) Trichlorido[28-acetyl-3-(tris-(hydroxylmethyl)amino-ethane)betulinic ester-κN]platinum(II): Synthesis and In Vitro Antitumor Activity. INORGANICS 2017. [DOI: 10.3390/inorganics5030056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
18
|
Gu HF, Li HZ, Xie XJ, Tang YL, Tang XQ, Nie YX, Liao DF. Oxidized low-density lipoprotein induced mouse hippocampal HT-22 cell damage via promoting the shift from autophagy to apoptosis. CNS Neurosci Ther 2017; 23:341-349. [PMID: 28233453 DOI: 10.1111/cns.12680] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/16/2017] [Accepted: 01/23/2017] [Indexed: 12/18/2022] Open
Abstract
AIMS Although oxidized low-density lipoprotein (ox-LDL) in the brain induces neuronal death, the mechanism underlying the damage effects remains largely unknown. Given that the ultimate outcome of a cell is depended on the balance between autophagy and apoptosis, this study was performed to explore whether ox-LDL induced HT-22 neuronal cell damage via autophagy impairment and apoptosis enhancement. METHODS Flow cytometry and transmission electron microscopy (TEM) were used to evaluate changes in cell apoptosis and autophagy, respectively. The protein expression of LC3-II, p62, Bcl-2, and Bax in HT-22 cells was measured by Western bolt analysis. RESULTS Our study confirmed that 100 μg/mL of ox-LDL not only promoted TH-22 cell apoptosis, characterized by elevated cell apoptosis rate and Bax protein expression, decreased Bcl-2 protein expression, and damaged cellular ultrastructures, but also impaired autophagy as indicated by the decreased LC3-II levels and the increased p62 levels. Importantly, all of these effects of ox-LDL were significantly aggravated by cotreatment with chloroquine (an inhibitor of autophagy flux). In contrast, cotreatment with rapamycin (an inducer of autophagy) remarkably reversed these effects of ox-LDL. CONCLUSIONS Taken together, our results indicated that ox-LDL-induced shift from autophagy to apoptosis contributes to HT-22 cell damage.
Collapse
Affiliation(s)
- Hong-Feng Gu
- Department of Physiology & Institute of Neuroscience, University of South China, Hengyang, China
| | - Hai-Zhe Li
- Department of Neurology of the First Affiliated Hospital, University of South China, Hengyang, China
| | - Xue-Jiao Xie
- Division of Stem Cell Regulation and Application, State Key Laboratory of Chinese Medicine Powder and Medicine Innovation in Hunan, Hunan University of Chinese Medicine, Changsha, China
| | - Ya-Ling Tang
- Department of Physiology & Institute of Neuroscience, University of South China, Hengyang, China
| | - Xiao-Qing Tang
- Department of Physiology & Institute of Neuroscience, University of South China, Hengyang, China
| | - Ya-Xiong Nie
- Department of Neurology of the First Affiliated Hospital, University of South China, Hengyang, China
| | - Duan-Fang Liao
- Division of Stem Cell Regulation and Application, State Key Laboratory of Chinese Medicine Powder and Medicine Innovation in Hunan, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
19
|
Hambright HG, Ghosh R. Autophagy: In the cROSshairs of cancer. Biochem Pharmacol 2016; 126:13-22. [PMID: 27789215 DOI: 10.1016/j.bcp.2016.10.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 10/21/2016] [Indexed: 12/18/2022]
Abstract
Two prominent features of tumors that contribute to oncogenic survival signaling are redox disruption, or oxidative stress phenotype, and high autophagy signaling, making both phenomena ideal therapeutic targets. However, the relationship between redox disruption and autophagy signaling is not well characterized and the clinical impact of reactive oxygen species (ROS)-generating chemotherapeutics on autophagy merits immediate attention as autophagy largely contributes to chemotherapeutic resistance. In this commentary we focus on melanoma, using it as an example to provide clarity to current literature regarding the roles of autophagy and redox signaling which can be applicable to initiation and maintenance of most tumor types. Further, we address the crosstalk between ROS and autophagy signaling during pharmacological intervention and cell fate decisions. We attempt to elucidate the role of autophagy in regulating cell fate following treatment with ROS-generating agents in preclinical and clinical settings and discuss the emerging role of autophagy in cell fate decisions and as a cell death mechanism. We also address technical aspects of redox and autophagy evaluation in experimental design and data interpretation. Lastly, we present a provocative view of the clinical relevance, emerging challenges in dual targeting of redox and autophagy pathways for therapy, and the future directions to be addressed in order to advance both basic and translational aspects of this field.
Collapse
Affiliation(s)
- Heather Graham Hambright
- Department of Urology, University of Texas Health Science Center at San Antonio, South Texas Research Facility Campus, 8403 Floyd Curl Drive, San Antonio, TX 78229, USA; Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, South Texas Research Facility Campus, 8403 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Rita Ghosh
- Department of Urology, University of Texas Health Science Center at San Antonio, South Texas Research Facility Campus, 8403 Floyd Curl Drive, San Antonio, TX 78229, USA; Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, South Texas Research Facility Campus, 8403 Floyd Curl Drive, San Antonio, TX 78229, USA; Department of Pharmacology, University of Texas Health Science Center at San Antonio, South Texas Research Facility Campus, 8403 Floyd Curl Drive, San Antonio, TX 78229, USA; Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, South Texas Research Facility Campus, 8403 Floyd Curl Drive, San Antonio, TX 78229, USA.
| |
Collapse
|