1
|
Liu Y, Tang Q, Tao Q, Dong H, Shi Z, Zhou L. Low-frequency magnetic field therapy for glioblastoma: Current advances, mechanisms, challenges and future perspectives. J Adv Res 2025; 69:531-543. [PMID: 38565404 PMCID: PMC11954840 DOI: 10.1016/j.jare.2024.03.024] [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: 12/28/2023] [Revised: 03/10/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common malignant tumour of the central nervous system. Despite recent advances in multimodal GBM therapy incorporating surgery, radiotherapy, systemic therapy (chemotherapy, targeted therapy), and supportive care, the overall survival (OS) remains poor, and long-term survival is rare. Currently, the primary obstacles hindering the effectiveness of GBM treatment are still the blood-brain barrier and tumor heterogeneity. In light of its substantial advantages over conventional therapies, such as strong penetrative ability and minimal side effects, low-frequency magnetic fields (LF-MFs) therapy has gradually caught the attention of scientists. AIM OF REVIEW In this review, we shed the light on the current status of applying LF-MFs in the treatment of GBM. We specifically emphasize our current understanding of the mechanisms by which LF-MFs mediate anticancer effects and the challenges faced by LF-MFs in treating GBM cells. Furthermore, we discuss the prospective applications of magnetic field therapy in the future treatment of GBM. Key scientific concepts of review: The review explores the current progress on the use of LF-MFs in the treatment of GBM with a special focus on the potential underlying mechanisms of LF-MFs in anticancer effects. Additionally, we also discussed the complex magnetic field features and biological characteristics related to magnetic bioeffects. Finally, we proposed a promising magnetic field treatment strategy for future applications in GBM therapy.
Collapse
Affiliation(s)
- Yinlong Liu
- Department of Neurosurgery, Huashan Hospital, Fudan University, China
| | - Qisheng Tang
- Department of Neurosurgery, Huashan Hospital, Fudan University, China; National Center for Neurological Disorders, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, China; Neurosurgical Institute of Fudan University, Shanghai, China; Shanghai Clinical Medical Center of Neurosurgery, China
| | - Quan Tao
- Shanghai Institute of Microsystem and Information Technology, China
| | - Hui Dong
- Shanghai Institute of Microsystem and Information Technology, China
| | - Zhifeng Shi
- Department of Neurosurgery, Huashan Hospital, Fudan University, China; National Center for Neurological Disorders, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, China; Neurosurgical Institute of Fudan University, Shanghai, China; Shanghai Clinical Medical Center of Neurosurgery, China.
| | - Liangfu Zhou
- Department of Neurosurgery, Huashan Hospital, Fudan University, China; National Center for Neurological Disorders, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, China; Neurosurgical Institute of Fudan University, Shanghai, China; Shanghai Clinical Medical Center of Neurosurgery, China.
| |
Collapse
|
2
|
Sharma A, Raut SS, Shukla A, Gupta S, Singh A, Mishra A. DDX3X dynamics, glioblastoma's genetic landscape, therapeutic advances, and autophagic interplay. Med Oncol 2024; 41:258. [PMID: 39368002 DOI: 10.1007/s12032-024-02525-z] [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: 06/18/2024] [Accepted: 09/23/2024] [Indexed: 10/07/2024]
Abstract
Glioblastoma is one of the most aggressive and deadly forms of cancer, posing significant challenges for the medical community. This review focuses on key aspects of Glioblastoma, including its genetic differences between primary and secondary types. Temozolomide is a major first-line treatment for Glioblastoma, and this article explores its development, how it works, and the issue of resistance that limits its effectiveness, prompting the need for new treatment strategies. Gene expression profiling has greatly advanced cancer research by revealing the molecular mechanisms of tumors, which is essential for creating targeted therapies for Glioblastoma. One important protein in this context is DDX3X, which plays various roles in cancer, sometimes promoting it or otherwise suppressing it. Additionally, autophagy, a process that maintains cellular balance, has complex implications in cancer treatment. Understanding autophagy helps to identify resistance mechanisms and potential treatments, with Chloroquine showing promise in treating Glioblastoma. This review covers the interplay between Glioblastoma, DDX3X, and autophagy, highlighting the challenges and potential strategies in treating this severe disease.
Collapse
Affiliation(s)
- Arpit Sharma
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Shruti S Raut
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Alok Shukla
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Shivani Gupta
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Amit Singh
- Department of Pharmacology, IMS-Banaras Hindu University, Varanasi, 221005, India.
| | - Abha Mishra
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India.
| |
Collapse
|
3
|
Elshazly AM, Xu J, Melhem N, Abdulnaby A, Elzahed AA, Saleh T, Gewirtz DA. Is Autophagy Targeting a Valid Adjuvant Strategy in Conjunction with Tyrosine Kinase Inhibitors? Cancers (Basel) 2024; 16:2989. [PMID: 39272847 PMCID: PMC11394573 DOI: 10.3390/cancers16172989] [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: 07/09/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Tyrosine kinase inhibitors (TKIs) represent a relatively large class of small-molecule inhibitors that compete with ATP for the catalytic binding site of tyrosine kinase proteins. While TKIs have demonstrated effectiveness in the treatment of multiple malignancies, including chronic myelogenous leukemia, gastrointestinal tumors, non-small cell lung cancers, and HER2-overexpressing breast cancers, as is almost always the case with anti-neoplastic agents, the development of resistance often imposes a limit on drug efficacy. One common survival response utilized by tumor cells to ensure their survival in response to different stressors, including anti-neoplastic drugs, is that of autophagy. The autophagic machinery in response to TKIs in multiple tumor models has largely been shown to be cytoprotective in nature, although there are a number of cases where autophagy has demonstrated a cytotoxic function. In this review, we provide an overview of the literature examining the role that autophagy plays in response to TKIs in different preclinical tumor model systems in an effort to determine whether autophagy suppression or modulation could be an effective adjuvant strategy to increase efficiency and/or overcome resistance to TKIs.
Collapse
Affiliation(s)
- Ahmed M. Elshazly
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, 401 College St., Richmond, VA 23298, USA;
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Jingwen Xu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China;
| | - Nebras Melhem
- Department of Anatomy, Physiology and Biochemistry, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan;
| | - Alsayed Abdulnaby
- Department of Pharmacognosy, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Aya A. Elzahed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Tareq Saleh
- Department of Pharmacology and Public Health, Faculty of Medicine, Hashemite University, Zarqa 13133, Jordan;
| | - David A. Gewirtz
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, 401 College St., Richmond, VA 23298, USA;
| |
Collapse
|
4
|
Mafi A, Khoshnazar SM, Shahpar A, Nabavi N, Hedayati N, Alimohammadi M, Hashemi M, Taheriazam A, Farahani N. Mechanistic insights into circRNA-mediated regulation of PI3K signaling pathway in glioma progression. Pathol Res Pract 2024; 260:155442. [PMID: 38991456 DOI: 10.1016/j.prp.2024.155442] [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: 04/11/2024] [Revised: 06/22/2024] [Accepted: 06/28/2024] [Indexed: 07/13/2024]
Abstract
Circular RNAs (CircRNAs) are non-coding RNAs (ncRNAs) characterized by a stable circular structure that regulates gene expression at both transcriptional and post-transcriptional levels. They play diverse roles, including protein interactions, DNA methylation modification, protein-coding potential, pseudogene creation, and miRNA sponging, all of which influence various physiological processes. CircRNAs are often highly expressed in brain tissues, and their levels vary with neural development, suggesting their significance in nervous system diseases such as gliomas. Research has shown that circRNA expression related to the PI3K pathway correlates with various clinical features of gliomas. There is an interact between circRNAs and the PI3K pathway to regulate glioma cell processes such as proliferation, differentiation, apoptosis, inflammation, angiogenesis, and treatment resistance. Additionally, PI3K pathway-associated circRNAs hold potential as biomarkers for cancer diagnosis, prognosis, and treatment. In this study, we reviewed the latest advances in the expression and cellular roles of PI3K-mediated circRNAs and their connections to glioma carcinogenesis and progression. We also highlighted the significance of circRNAs as diagnostic and prognostic biomarkers and therapeutic targets in glioma.
Collapse
Affiliation(s)
- Alireza Mafi
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyedeh Mahdieh Khoshnazar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Amirhossein Shahpar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, Canada
| | - Neda Hedayati
- School of Medicine, Iran University of Medical Science, Tehran, Iran.
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Najma Farahani
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
5
|
Simpson JE, Muir MT, Lee M, Naughton C, Gilbert N, Pollard SM, Gammoh N. Autophagy supports PDGFRA-dependent brain tumor development by enhancing oncogenic signaling. Dev Cell 2024; 59:228-243.e7. [PMID: 38113891 DOI: 10.1016/j.devcel.2023.11.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 07/29/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023]
Abstract
Autophagy is a conserved cellular degradation process. While autophagy-related proteins were shown to influence the signaling and trafficking of some receptor tyrosine kinases, the relevance of this during cancer development is unclear. Here, we identify a role for autophagy in regulating platelet-derived growth factor receptor alpha (PDGFRA) signaling and levels. We find that PDGFRA can be targeted for autophagic degradation through the activity of the autophagy cargo receptor p62. As a result, short-term autophagy inhibition leads to elevated levels of PDGFRA but an unexpected defect in PDGFA-mediated signaling due to perturbed receptor trafficking. Defective PDGFRA signaling led to its reduced levels during prolonged autophagy inhibition, suggesting a mechanism of adaptation. Importantly, PDGFA-driven gliomagenesis in mice was disrupted when autophagy was inhibited in a manner dependent on Pten status, thus highlighting a genotype-specific role for autophagy during tumorigenesis. In summary, our data provide a mechanism by which cells require autophagy to drive tumor formation.
Collapse
Affiliation(s)
- Joanne E Simpson
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK
| | - Morwenna T Muir
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK
| | - Martin Lee
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK
| | - Catherine Naughton
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XU, UK
| | - Nick Gilbert
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XU, UK
| | - Steven M Pollard
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK; Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Noor Gammoh
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK.
| |
Collapse
|
6
|
Qin Y, Xiong S, Ren J, Sethi G. Autophagy machinery in glioblastoma: The prospect of cell death crosstalk and drug resistance with bioinformatics analysis. Cancer Lett 2024; 580:216482. [PMID: 37977349 DOI: 10.1016/j.canlet.2023.216482] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/01/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
Brain tumors are common malignancies with high mortality and morbidity in which glioblastoma (GB) is a grade IV astrocytoma with heterogeneous nature. The conventional therapeutics for the GB mainly include surgery and chemotherapy, however their efficacy has been compromised due to the aggressiveness of tumor cells. The dysregulation of cell death mechanisms, especially autophagy has been reported as a factor causing difficulties in cancer therapy. As a mechanism contributing to cell homeostasis, the autophagy process is hijacked by tumor cells for the purpose of aggravating cancer progression and drug resistance. The autophagy function is context-dependent and its role can be lethal or protective in cancer. The aim of the current paper is to highlight the role of autophagy in the regulation of GB progression. The cytotoxic function of autophagy can promote apoptosis and ferroptosis in GB cells and vice versa. Autophagy dysregulation can cause drug resistance and radioresistance in GB. Moreover, stemness can be regulated by autophagy and overall growth as well as metastasis are affected by autophagy. The various interventions including administration of synthetic/natural products and nanoplatforms can target autophagy. Therefore, autophagy can act as a promising target in GB therapy.
Collapse
Affiliation(s)
- Yi Qin
- Department of Lab, Chifeng Cancer Hospital (The 2nd Afflicted Hospital of Chifeng University), Chifeng University, Chifeng City, Inner Mongolia Autonomous Region, 024000, China.
| | - Shengjun Xiong
- Department of Cardiology, Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jun Ren
- Department of Cardiology, Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Gautam Sethi
- Department of Pharmacology, National University of Singapore, NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, 16 Medical Drive, Singapore, 117600, Singapore.
| |
Collapse
|
7
|
Gustafson DL, Viola LO, Towers CG, Das S, Duval DL, Van Eaton KM. Sensitivity of osteosarcoma cell lines to autophagy inhibition as determined by pharmacologic and genetic manipulation. Vet Comp Oncol 2023; 21:726-738. [PMID: 37724007 PMCID: PMC11470750 DOI: 10.1111/vco.12937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/05/2023] [Accepted: 09/05/2023] [Indexed: 09/20/2023]
Abstract
Pharmacologic inhibition of autophagy can be achieved using lysosomotropic agents such as hydroxychloroquine (HCQ) that interfere with fusion of the autophagosome to the lysosome thus preventing completion of the recycling process. The goal of the present study is to determine the sensitivity of eight canine (cOSA) and four human (hOSA) osteosarcoma tumour cell lines to antiproliferative and cytotoxic effects of lysosomal autophagy inhibitors, and to compare these results to the autophagy-dependence measured using a CRISPR/Cas9 live-cell imaging assay in OSA and other tumour cell lines. Antiproliferative and cytotoxic response to HCQ and Lys05 was determined using live cell imaging and YOYO-1 staining. CRISPR/Cas9 live cell imaging screen was done using species specific guide RNA's and transfection of reagents into cells. Response to autophagy core genes was compared to response to an essential (PCNA) and non-essential (FOXO3A) gene. cOSA and hOSA cell lines showed similar antiproliferative and cytotoxic responses to HCQ and Lys05 with median lethal dose (Dm ) values ranging from 4.6-15.8 μM and 2.1-5.1 μM for measures of anti-proliferative response, respectively. A relationship was observed between antiproliferative responses to HCQ and Lys05 and VPS34 CRISPR score with Dm values correlating with VPS34 response (r = 0.968 and 0.887) in a species independent manner. The results show that a subset of cOSA and hOSA cell lines are autophagy-dependent and sensitive to HCQ at pharmacologically-relevant exposures.
Collapse
Affiliation(s)
- Daniel L. Gustafson
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, USA
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
- Developmental Therapeutics Program, University of Colorado Cancer Center, Aurora, Colorado, USA
| | - Lindsey O. Viola
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
| | - Christina G. Towers
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Sciences, La Jolla, California, USA
| | - Sunetra Das
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
| | - Dawn L. Duval
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
- Developmental Therapeutics Program, University of Colorado Cancer Center, Aurora, Colorado, USA
| | - Kristen M. Van Eaton
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
8
|
Pizzimenti C, Fiorentino V, Franchina M, Martini M, Giuffrè G, Lentini M, Silvestris N, Di Pietro M, Fadda G, Tuccari G, Ieni A. Autophagic-Related Proteins in Brain Gliomas: Role, Mechanisms, and Targeting Agents. Cancers (Basel) 2023; 15:cancers15092622. [PMID: 37174088 PMCID: PMC10177137 DOI: 10.3390/cancers15092622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
The present review focuses on the phenomenon of autophagy, a catabolic cellular process, which allows for the recycling of damaged organelles, macromolecules, and misfolded proteins. The different steps able to activate autophagy start with the formation of the autophagosome, mainly controlled by the action of several autophagy-related proteins. It is remarkable that autophagy may exert a double role as a tumour promoter and a tumour suppressor. Herein, we analyse the molecular mechanisms as well as the regulatory pathways of autophagy, mainly addressing their involvement in human astrocytic neoplasms. Moreover, the relationships between autophagy, the tumour immune microenvironment, and glioma stem cells are discussed. Finally, an excursus concerning autophagy-targeting agents is included in the present review in order to obtain additional information for the better treatment and management of therapy-resistant patients.
Collapse
Affiliation(s)
- Cristina Pizzimenti
- Translational Molecular Medicine and Surgery, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy
| | - Vincenzo Fiorentino
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| | - Mariausilia Franchina
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| | - Maurizio Martini
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| | - Giuseppe Giuffrè
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| | - Maria Lentini
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| | - Nicola Silvestris
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Oncology Section, University of Messina, 98125 Messina, Italy
| | - Martina Di Pietro
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Oncology Section, University of Messina, 98125 Messina, Italy
| | - Guido Fadda
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| | - Giovanni Tuccari
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| | - Antonio Ieni
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| |
Collapse
|
9
|
Zhang Z, Chen WQ, Zhang SQ, Bai JX, Liu B, Yung KKL, Ko JKS. Isoliquiritigenin inhibits pancreatic cancer progression through blockade of p38 MAPK-regulated autophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 106:154406. [PMID: 36029643 DOI: 10.1016/j.phymed.2022.154406] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/22/2022] [Accepted: 08/17/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Pancreatic cancer has been characterized by poor prognosis, early metastasis and dissatisfactory treatment outcome. The high basal level of autophagy in tumor cells leads to chemoresistance and tumor progression. Thus, it is imminent to explore novel effective chemotherapeutic adjuvants to increase patients' survival rate. Isoliquiritigenin (ISL) is a bioactive flavonoid obtained from the Traditional Chinese herbal medicine Glycyrrhiza glabra, and it possesses a broad range of pharmacological effects. In this study, the anti-cancer effect of ISL in pancreatic cancer treatment and the underlying mechanism are investigated. METHODS MTT assay, colony formation and EdU analysis were performed to explore the growth inhibition of ISL on pancreatic cancer cells. Apoptosis were analyzed using TUNEL and flow cytometry. The formations of autophagosomes were analyzed by immunofluorescence microscopy and transmission electron microscopy. RFP-GFP-LC3B probe was applied to detect the autophagy flux. To assess the structural interaction of ISL with p38 protein, molecular docking assays were performed. The molecular mechanism was elucidated by using western immunoblotting. Subsequently, the inhibition of ISL on tumor growth was determined in vivo using pancreatic tumor mice model. RESULTS ISL inhibited pancreatic cancer cell growth and induced apoptosis, both in vitro and in vivo. ISL caused accumulation of autophagosome through blockade of late stage autophagic flux. Moreover, autophagy inducer rapamycin enhanced ISL-evoked cell growth inhibition and promoted apoptosis, while inhibition of autophagosome formation by siAtg5 attenuated ISL-induced apoptosis. It is remarkable that ISL synergistically sensitized the cytotoxic effect of gemcitabine and 5-fluorouracil on pancreatic cancer cells as both drugs induced autophagy. Molecular docking analysis has indicated that ISL acted by direct targeting of p38 MAPK, which was confirmed by ISL-induced phosphorylation of p38. The autophagy flux induced by p38 inhibitor SB203580 was blocked by ISL, with further increasing toxicity of ISL in pancreatic cancer cells. CONCLUSION The results have revealed that ISL inhibited pancreatic cancer progression by blockade of autophagy through p38 MAPK signaling.
Collapse
Affiliation(s)
- Zhu Zhang
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; Department of Biology, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong SAR, China; Golden Meditech Centre for NeuroRegeneration Sciences, Hong Kong Baptist University, Hong Kong SAR, China
| | - Wen-Qing Chen
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; Department of Biology, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong SAR, China
| | - Shi-Qing Zhang
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jing-Xuan Bai
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Bin Liu
- Department of Traditional Chinese Medicine, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Ken Kin-Lam Yung
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong SAR, China; Golden Meditech Centre for NeuroRegeneration Sciences, Hong Kong Baptist University, Hong Kong SAR, China.
| | - Joshua Ka-Shun Ko
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Hong Kong SAR, China.
| |
Collapse
|
10
|
Jovanović Stojanov S, Kostić A, Ljujić M, Lupšić E, Schenone S, Pešić M, Dinić J. Autophagy Inhibition Enhances Anti-Glioblastoma Effects of Pyrazolo[3,4-d]pyrimidine Tyrosine Kinase Inhibitors. Life (Basel) 2022; 12:life12101503. [PMID: 36294938 PMCID: PMC9605466 DOI: 10.3390/life12101503] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/14/2022] [Accepted: 09/23/2022] [Indexed: 01/18/2023] Open
Abstract
Drug resistance presents a major obstacle to the successful treatment of glioblastoma. Autophagy plays a key role in drug resistance, particularly in relation to targeted therapy, which has prompted the use of autophagy inhibitors to increase the effectiveness of targeted therapeutics. The ability of two Src tyrosine kinase inhibitors, Si306 and its prodrug pro-Si306, to induce autophagy was evaluated in the human glioblastoma cell line U87 and its multidrug-resistant counterpart U87-TxR. Autophagy markers were assessed by flow cytometry, microscopy, and Western blot, and induction of autophagy by these compounds was demonstrated after 3 h as well as 48 h. The effects of Si306 and pro-Si306 on cell proliferation and cell death were examined in the presence or absence of autophagy inhibition by bafilomycin A1. Combined treatments of Si306 and pro-Si306 with bafilomycin A1 were synergistic in nature, and the inhibition of autophagy sensitized glioblastoma cells to Src tyrosine kinase inhibitors. Si306 and pro-Si306 more strongly inhibited cell proliferation and triggered necrosis in combination with bafilomycin A1. Our findings suggest that modulation of Si306- and pro-Si306-induced autophagy can be used to enhance the anticancer effects of these Src tyrosine kinase inhibitors and overcome the drug-resistant phenotype in glioblastoma cells.
Collapse
Affiliation(s)
- Sofija Jovanović Stojanov
- Department of Neurobiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Ana Kostić
- Department of Neurobiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Mila Ljujić
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia
| | - Ema Lupšić
- Department of Neurobiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Silvia Schenone
- Department of Pharmacy, University of Genova, Viale Benedetto XV 3, 16132 Genova, Italy
| | - Milica Pešić
- Department of Neurobiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Jelena Dinić
- Department of Neurobiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
- Correspondence: ; Tel.: +381-112078406
| |
Collapse
|
11
|
Zhao A, Liu N, Yao M, Zhang Y, Yao Z, Feng Y, Liu J, Zhou G. A Review of Neuroprotective Effects and Mechanisms of Ginsenosides From Panax Ginseng in Treating Ischemic Stroke. Front Pharmacol 2022; 13:946752. [PMID: 35873557 PMCID: PMC9302711 DOI: 10.3389/fphar.2022.946752] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/14/2022] [Indexed: 12/14/2022] Open
Abstract
Ischemic stroke has been considered one of the leading causes of mortality and disability worldwide, associated with a series of complex pathophysiological processes. However, effective therapeutic methods for ischemic stroke are still limited. Panax ginseng, a valuable traditional Chinese medicine, has been long used in eastern countries for various diseases. Ginsenosides, the main active ingredient of Panax ginseng, has demonstrated neuroprotective effects on ischemic stroke injury during the last decade. In this article, we summarized the pathophysiology of ischemic stroke and reviewed the literature on ginsenosides studies in preclinical and clinical ischemic stroke. Available findings showed that both major ginsenosides and minor ginsenosides (such as Rg3, Rg5, and Rh2) has a potential neuroprotective effect, mainly through attenuating the excitotoxicity, Ca2+ overload, mitochondria dysfunction, blood-brain barrier (BBB) permeability, anti-inflammation, anti-oxidative, anti-apoptosis, anti-pyroptosis, anti-autophagy, improving angiogenesis, and neurogenesis. Therefore, this review brings a current understanding of the mechanisms of ginsenosides in the treatment of ischemic stroke. Further studies, especially in clinical trials, will be important to confirm the clinical value of ginseng and ginsenosides.
Collapse
Affiliation(s)
- Aimei Zhao
- Department of Acupuncture and Moxibustion, Neuroscience Centre, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Nan Liu
- Beijing Increasepharm Safety and Efficacy Co., Ltd., Beijing, China
| | - Mingjiang Yao
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yehao Zhang
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Zengyu Yao
- Department of Acupuncture and Moxibustion, Neuroscience Centre, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yujing Feng
- Department of Anesthesiology, Punan Hospital, Shanghai, China
| | - Jianxun Liu
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Jianxun Liu, ; Guoping Zhou,
| | - Guoping Zhou
- Department of Acupuncture and Moxibustion, Neuroscience Centre, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- *Correspondence: Jianxun Liu, ; Guoping Zhou,
| |
Collapse
|
12
|
Ghaznavi H, Shirvaliloo M, Zarebkohan A, Shams Z, Radnia F, Bahmanpour Z, Sargazi S, Saravani R, Shirvalilou S, Shahraki O, Shahraki S, Nazarlou Z, Sheervalilou R. An Updated Review on Implications of Autophagy and Apoptosis in Tumorigenesis: Possible Alterations in Autophagy through Engineered Nanomaterials and Their Importance in Cancer Therapy. Mol Pharmacol 2021; 100:119-143. [PMID: 33990406 DOI: 10.1124/molpharm.121.000234] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/19/2021] [Indexed: 12/17/2022] Open
Abstract
Most commonly recognized as a catabolic pathway, autophagy is a perplexing mechanism through which a living cell can free itself of excess cytoplasmic components, i.e., organelles, by means of certain membranous vesicles or lysosomes filled with degrading enzymes. Upon exposure to external insult or internal stimuli, the cell might opt to activate such a pathway, through which it can gain control over the maintenance of intracellular components and thus sustain homeostasis by intercepting the formation of unnecessary structures or eliminating the already present dysfunctional or inutile organelles. Despite such appropriateness, autophagy might also be considered a frailty for the cell, as it has been said to have a rather complicated role in tumorigenesis. A merit in the early stages of tumor formation, autophagy appears to be salutary because of its tumor-suppressing effects. In fact, several investigations on tumorigenesis have reported diminished levels of autophagic activity in tumor cells, which might result in transition to malignancy. On the contrary, autophagy has been suggested to be a seemingly favorable mechanism to progressed malignancies, as it contributes to survival of such cells. Based on the recent literature, this mechanism might also be activated upon the entry of engineered nanomaterials inside a cell, supposedly protecting the host from foreign materials. Accordingly, there is a good chance that therapeutic interventions for modulating autophagy in malignant cells using nanoparticles may sensitize cancerous cells to certain treatment modalities, e.g., radiotherapy. In this review, we will discuss the signaling pathways involved in autophagy and the significance of the mechanism itself in apoptosis and tumorigenesis while shedding light on possible alterations in autophagy through engineered nanomaterials and their potential therapeutic applications in cancer. SIGNIFICANCE STATEMENT: Autophagy has been said to have a complicated role in tumorigenesis. In the early stages of tumor formation, autophagy appears to be salutary because of its tumor-suppressing effects. On the contrary, autophagy has been suggested to be a favorable mechanism to progressed malignancies. This mechanism might be affected upon the entry of nanomaterials inside a cell. Accordingly, therapeutic interventions for modulating autophagy using nanoparticles may sensitize cancerous cells to certain therapies.
Collapse
Affiliation(s)
- Habib Ghaznavi
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Milad Shirvaliloo
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Amir Zarebkohan
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Zinat Shams
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Fatemeh Radnia
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Zahra Bahmanpour
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Saman Sargazi
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Ramin Saravani
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Sakine Shirvalilou
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Omolbanin Shahraki
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Sheida Shahraki
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Ziba Nazarlou
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Roghayeh Sheervalilou
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| |
Collapse
|
13
|
Batara DCR, Choi MC, Shin HU, Kim H, Kim SH. Friend or Foe: Paradoxical Roles of Autophagy in Gliomagenesis. Cells 2021; 10:1411. [PMID: 34204169 PMCID: PMC8227518 DOI: 10.3390/cells10061411] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/30/2021] [Accepted: 06/03/2021] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive type of primary brain tumor in adults, with a poor median survival of approximately 15 months after diagnosis. Despite several decades of intensive research on its cancer biology, treatment for GBM remains a challenge. Autophagy, a fundamental homeostatic mechanism, is responsible for degrading and recycling damaged or defective cellular components. It plays a paradoxical role in GBM by either promoting or suppressing tumor growth depending on the cellular context. A thorough understanding of autophagy's pleiotropic roles is needed to develop potential therapeutic strategies for GBM. In this paper, we discussed molecular mechanisms and biphasic functions of autophagy in gliomagenesis. We also provided a summary of treatments for GBM, emphasizing the importance of autophagy as a promising molecular target for treating GBM.
Collapse
Affiliation(s)
- Don Carlo Ramos Batara
- Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju 61186, Korea; (D.C.R.B.); (H.-U.S.)
| | - Moon-Chang Choi
- Department of Biomedical Science, Chosun University, Gwangju 61452, Korea;
| | - Hyeon-Uk Shin
- Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju 61186, Korea; (D.C.R.B.); (H.-U.S.)
| | - Hyunggee Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea;
| | - Sung-Hak Kim
- Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju 61186, Korea; (D.C.R.B.); (H.-U.S.)
| |
Collapse
|
14
|
Hou X, Han L, An B, Cai J. Autophagy induced by Vip3Aa has a pro-survival role in Spodoptera frugiperda Sf9 cells. Virulence 2021; 12:509-519. [PMID: 33509041 PMCID: PMC7849784 DOI: 10.1080/21505594.2021.1878747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Vip3Aa is an insecticidal protein that can effectively control certain lepidopteran pests and has been used widely in biological control. However, the mechanism of action of Vip3Aa is unclear. In the present study, we showed that Vip3Aa could cause autophagy in Sf9 cells, which was confirmed by the increased numbers of GFP-Atg8 puncta, the appearance of autophagic vacuoles, and an elevated Atg8-II protein level. Moreover, we found that the AMPK-mTOR-ULK1 pathway is involved in Vip3Aa-induced autophagy, which might be associated with the destruction of ATP homeostasis in Vip3Aa-treated cells. Both the elevated p62 level and the increased numbers of GFP-RFP-Atg8 yellow fluorescent spots demonstrated that autophagy in Sf9 cells was inhibited at 24 h after Vip3Aa treatment. With the prolongation of Vip3Aa treatment time, this inhibition became more serious and led to autophagosome accumulation. Genetic knockdown of ATG5 or the use of the autophagy inhibitor 3-MA further increased the sensitivity of Sf9 cells to Vip3Aa. Overexpression of ATG5 reduced the cell mortality of Vip3Aa-treated cells. In summary, the results revealed that autophagy induced by Vip3Aa has a pro-survival role, which might be related to the development of insect resistance.
Collapse
Affiliation(s)
- Xiaoyue Hou
- Department of Microbiology, College of Life Sciences, Nankai University , Tianjin, China.,Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University , Lianyungang, China.,College of Food Science and Engineering, Jiangsu Ocean University , Lianyungang, China
| | - Lu Han
- Department of Microbiology, College of Life Sciences, Nankai University , Tianjin, China
| | - Baoju An
- Department of Microbiology, College of Life Sciences, Nankai University , Tianjin, China
| | - Jun Cai
- Department of Microbiology, College of Life Sciences, Nankai University , Tianjin, China.,Key Laboratory of Molecular Microbiology and Technology, Ministry of Education , Tianjin, China.,Tianjin Key Laboratory of Microbial Functional Genomics , Tianjin, China
| |
Collapse
|
15
|
Khan I, Baig MH, Mahfooz S, Rahim M, Karacam B, Elbasan EB, Ulasov I, Dong JJ, Hatiboglu MA. Deciphering the Role of Autophagy in Treatment of Resistance Mechanisms in Glioblastoma. Int J Mol Sci 2021; 22:ijms22031318. [PMID: 33525678 PMCID: PMC7865981 DOI: 10.3390/ijms22031318] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a process essential for cellular energy consumption, survival, and defense mechanisms. The role of autophagy in several types of human cancers has been explicitly explained; however, the underlying molecular mechanism of autophagy in glioblastoma remains ambiguous. Autophagy is thought to be a “double-edged sword”, and its effect on tumorigenesis varies with cell type. On the other hand, autophagy may play a significant role in the resistance mechanisms against various therapies. Therefore, it is of the utmost importance to gain insight into the molecular mechanisms deriving the autophagy-mediated therapeutic resistance and designing improved treatment strategies for glioblastoma. In this review, we discuss autophagy mechanisms, specifically its pro-survival and growth-suppressing mechanisms in glioblastomas. In addition, we try to shed some light on the autophagy-mediated activation of the cellular mechanisms supporting radioresistance and chemoresistance in glioblastoma. This review also highlights autophagy’s involvement in glioma stem cell behavior, underlining its role as a potential molecular target for therapeutic interventions.
Collapse
Affiliation(s)
- Imran Khan
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy Mahallesi, Beykoz, 34820 Istanbul, Turkey; (I.K.); (S.M.); (B.K.)
| | - Mohammad Hassan Baig
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea;
| | - Sadaf Mahfooz
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy Mahallesi, Beykoz, 34820 Istanbul, Turkey; (I.K.); (S.M.); (B.K.)
| | - Moniba Rahim
- Department of Biosciences, Integral University, Lucknow, Uttar Pradesh 226026, India;
| | - Busra Karacam
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy Mahallesi, Beykoz, 34820 Istanbul, Turkey; (I.K.); (S.M.); (B.K.)
| | - Elif Burce Elbasan
- Department of Neurosurgery, Bezmialem Vakif University Medical School, Vatan Street, Fatih, 34093 Istanbul, Turkey;
| | - Ilya Ulasov
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Jae-June Dong
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea;
- Correspondence: (J.-J.D.); (M.A.H.)
| | - Mustafa Aziz Hatiboglu
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy Mahallesi, Beykoz, 34820 Istanbul, Turkey; (I.K.); (S.M.); (B.K.)
- Department of Neurosurgery, Bezmialem Vakif University Medical School, Vatan Street, Fatih, 34093 Istanbul, Turkey;
- Correspondence: (J.-J.D.); (M.A.H.)
| |
Collapse
|
16
|
Shao CS, Zhou XH, Zheng XX, Huang Q. Ganoderic acid D induces synergistic autophagic cell death except for apoptosis in ESCC cells. JOURNAL OF ETHNOPHARMACOLOGY 2020; 262:113213. [PMID: 32755651 DOI: 10.1016/j.jep.2020.113213] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/12/2020] [Accepted: 07/22/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHAMACOLOGICAL RELEVANCE Ganoderma lucidum has been used as a medicinal mushroom for more than 2000 years in China. Ganoderic acid D (GAD) as a representative active triterpenoid from Ganoderma lucidum is known to possess anticancer activity. However, the mechanism involved in its anticancer cell process is still largely elusive. AIM OF THE STUDY Our study aimed to investigate the anticancer effects of GAD on the esophageal squamous cell carcinoma (ESCC) cells and the underlying mechanisms at the cell level. MATERIALS AND METHODS EC9706 and Eca109 cells were treated with GAD (0, 10, 20, 40 μM) for 24 h. The cell viability, cell cycle, reactive oxygen species (ROS), mitochondrial membrane potential (MMP), apoptosis rate, caspase-3 activity, autophagic flux, lysosomal function were examined. Cell cycle, apoptotic, autophagy and mTOR signal pathway related proteins such as P53, Cyclin B1, CytoC, PARP, Beclin-1, P62, LC3, PI3K, AKT and mTOR were analyzed by Western blot approach. RESULTS GAD inhibited cell proliferation and induced both apoptosis and autophagic cell death. In particular, we found that in the early stage of the autophagic process, GAD could initiate and enhance the autophagy signal while in the late stage it on the contrary could block the autophagic flux by impairing the autophagosome-lysosome fusion and inhibited the lysosomal degradation. Besides the autophagic cell death, GAD also induced the apoptosis mediated by caspase-related process in parallel. The mechanism involved for the synergistic apoptotic and autophagic cell death was also explored. We found that GAD down-regulated the expression of PI3K, AKT and mTOR phosphorylated proteins in the mTOR signaling pathway which thus led to the synergistic effect on apoptosis and autophagic cell death in the ESCC cells. CONCLUSIONS In summary, this study has documented that GAD may inhibit cell proliferation through the mTOR pathway in ESCC cells, and induce synergistic apoptosis and autophagic cell death by disrupting the autophagic flux. This work therefore also suggests that GAD may be used as an efficient anticancer adjuvant for ESCC cancer therapy.
Collapse
Affiliation(s)
- Chang-Sheng Shao
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; University of Science and Technology of China, Hefei, 230026, China.
| | - Xiu-Hong Zhou
- Center of Biology, Anhui Agricultural University, Hefei, China.
| | - Xin-Xin Zheng
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; University of Science and Technology of China, Hefei, 230026, China.
| | - Qing Huang
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; University of Science and Technology of China, Hefei, 230026, China.
| |
Collapse
|
17
|
Shnaider PV, Ivanova OM, Malyants IK, Anufrieva KS, Semenov IA, Pavlyukov MS, Lagarkova MA, Govorun VM, Shender VO. New Insights into Therapy-Induced Progression of Cancer. Int J Mol Sci 2020; 21:E7872. [PMID: 33114182 PMCID: PMC7660620 DOI: 10.3390/ijms21217872] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/19/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023] Open
Abstract
The malignant tumor is a complex heterogeneous set of cells functioning in a no less heterogeneous microenvironment. Like any dynamic system, cancerous tumors evolve and undergo changes in response to external influences, including therapy. Initially, most tumors are susceptible to treatment. However, remaining cancer cells may rapidly reestablish the tumor after a temporary remission. These new populations of malignant cells usually have increased resistance not only to the first-line agent, but also to the second- and third-line drugs, leading to a significant decrease in patient survival. Multiple studies describe the mechanism of acquired therapy resistance. In past decades, it became clear that, in addition to the simple selection of pre-existing resistant clones, therapy induces a highly complicated and tightly regulated molecular response that allows tumors to adapt to current and even subsequent therapeutic interventions. This review summarizes mechanisms of acquired resistance, such as secondary genetic alterations, impaired function of drug transporters, and autophagy. Moreover, we describe less obvious molecular aspects of therapy resistance in cancers, including epithelial-to-mesenchymal transition, cell cycle alterations, and the role of intercellular communication. Understanding these molecular mechanisms will be beneficial in finding novel therapeutic approaches for cancer therapy.
Collapse
Affiliation(s)
- Polina V. Shnaider
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow 119435, Russia; (P.V.S.); (O.M.I.); (K.S.A.); (M.A.L.)
- Laboratory of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow 119435, Russia; (I.K.M.); (I.A.S.)
- Faculty of Biology, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Olga M. Ivanova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow 119435, Russia; (P.V.S.); (O.M.I.); (K.S.A.); (M.A.L.)
- Laboratory of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow 119435, Russia; (I.K.M.); (I.A.S.)
| | - Irina K. Malyants
- Laboratory of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow 119435, Russia; (I.K.M.); (I.A.S.)
- Faculty of Chemical-Pharmaceutical Technologies and Biomedical Drugs, Mendeleev University of Chemical Technology of Russia, Moscow 125047, Russia
| | - Ksenia S. Anufrieva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow 119435, Russia; (P.V.S.); (O.M.I.); (K.S.A.); (M.A.L.)
- Laboratory of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow 119435, Russia; (I.K.M.); (I.A.S.)
- Moscow Institute of Physics and Technology (State University), Dolgoprudny 141701, Russia
| | - Ilya A. Semenov
- Laboratory of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow 119435, Russia; (I.K.M.); (I.A.S.)
| | - Marat S. Pavlyukov
- Laboratory of Membrane Bioenergetics, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia;
| | - Maria A. Lagarkova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow 119435, Russia; (P.V.S.); (O.M.I.); (K.S.A.); (M.A.L.)
- Laboratory of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow 119435, Russia; (I.K.M.); (I.A.S.)
| | - Vadim M. Govorun
- Laboratory of Simple Systems, Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow 119435, Russia;
| | - Victoria O. Shender
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow 119435, Russia; (P.V.S.); (O.M.I.); (K.S.A.); (M.A.L.)
- Laboratory of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow 119435, Russia; (I.K.M.); (I.A.S.)
- Laboratory of Molecular Oncology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia
| |
Collapse
|
18
|
Simpson JE, Gammoh N. The impact of autophagy during the development and survival of glioblastoma. Open Biol 2020; 10:200184. [PMID: 32873152 PMCID: PMC7536068 DOI: 10.1098/rsob.200184] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 07/30/2020] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma is the most common and aggressive adult brain tumour, with poor median survival and limited treatment options. Following surgical resection and chemotherapy, recurrence of the disease is inevitable. Genomic studies have identified key drivers of glioblastoma development, including amplifications of receptor tyrosine kinases, which drive tumour growth. To improve treatment, it is crucial to understand survival response processes in glioblastoma that fuel cell proliferation and promote resistance to treatment. One such process is autophagy, a catabolic pathway that delivers cellular components sequestered into vesicles for lysosomal degradation. Autophagy plays an important role in maintaining cellular homeostasis and is upregulated during stress conditions, such as limited nutrient and oxygen availability, and in response to anti-cancer therapy. Autophagy can also regulate pro-growth signalling and metabolic rewiring of cancer cells in order to support tumour growth. In this review, we will discuss our current understanding of how autophagy is implicated in glioblastoma development and survival. When appropriate, we will refer to findings derived from the role of autophagy in other cancer models and predict the outcome of manipulating autophagy during glioblastoma treatment.
Collapse
Affiliation(s)
| | - Noor Gammoh
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK
| |
Collapse
|
19
|
Dinić J, Efferth T, García-Sosa AT, Grahovac J, Padrón JM, Pajeva I, Rizzolio F, Saponara S, Spengler G, Tsakovska I. Repurposing old drugs to fight multidrug resistant cancers. Drug Resist Updat 2020; 52:100713. [PMID: 32615525 DOI: 10.1016/j.drup.2020.100713] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/04/2020] [Accepted: 06/06/2020] [Indexed: 02/08/2023]
Abstract
Overcoming multidrug resistance represents a major challenge for cancer treatment. In the search for new chemotherapeutics to treat malignant diseases, drug repurposing gained a tremendous interest during the past years. Repositioning candidates have often emerged through several stages of clinical drug development, and may even be marketed, thus attracting the attention and interest of pharmaceutical companies as well as regulatory agencies. Typically, drug repositioning has been serendipitous, using undesired side effects of small molecule drugs to exploit new disease indications. As bioinformatics gain increasing popularity as an integral component of drug discovery, more rational approaches are needed. Herein, we show some practical examples of in silico approaches such as pharmacophore modelling, as well as pharmacophore- and docking-based virtual screening for a fast and cost-effective repurposing of small molecule drugs against multidrug resistant cancers. We provide a timely and comprehensive overview of compounds with considerable potential to be repositioned for cancer therapeutics. These drugs are from diverse chemotherapeutic classes. We emphasize the scope and limitations of anthelmintics, antibiotics, antifungals, antivirals, antimalarials, antihypertensives, psychopharmaceuticals and antidiabetics that have shown extensive immunomodulatory, antiproliferative, pro-apoptotic, and antimetastatic potential. These drugs, either used alone or in combination with existing anticancer chemotherapeutics, represent strong candidates to prevent or overcome drug resistance. We particularly focus on outcomes and future perspectives of drug repositioning for the treatment of multidrug resistant tumors and discuss current possibilities and limitations of preclinical and clinical investigations.
Collapse
Affiliation(s)
- Jelena Dinić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | | | - Jelena Grahovac
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - José M Padrón
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González (IUBO AG), Universidad de La Laguna, Avda. Astrofísico Francisco Sánchez 2, E-38071 La Laguna, Spain.
| | - Ilza Pajeva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 105, 1113 Sofia, Bulgaria
| | - Flavio Rizzolio
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, 301724 Venezia-Mestre, Italy; Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Simona Saponara
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Gabriella Spengler
- Department of Medical Microbiology and Immunobiology, Faculty of Medicine, University of Szeged, H-6720 Szeged, Dóm tér 10, Hungary
| | - Ivanka Tsakovska
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 105, 1113 Sofia, Bulgaria
| |
Collapse
|
20
|
Agarwal S, Maekawa T. Nano delivery of natural substances as prospective autophagy modulators in glioblastoma. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 29:102270. [PMID: 32702467 DOI: 10.1016/j.nano.2020.102270] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 12/14/2022]
Abstract
Glioblastoma is the most destructive type of malignant brain tumor in humans due to cancer relapse. Latest studies have indicated that cancer cells are more reliant on autophagy for survival than non-cancer cells. Autophagy is entitled as programmed cell death type II and studies imply that it is a comeback of cancer cells to innumerable anti-cancer therapies. To diminish the adverse consequences of chemotherapeutics, numerous herbs of natural origin have been retained in cancer treatments. Additionally, autophagy induction occurs via their tumor suppressive actions that could cause cell senescence and increase apoptosis-independent cell death. However, most of the drugs have poor solubility and thus nano drug delivery systems possess excessive potential to improve the aqueous solubility and bioavailability of encapsulated drugs. There is a pronounced need for more therapies for glioblastoma treatment and hereby, the fundamental mechanisms of natural autophagy modulators in glioblastoma are prudently reviewed in this article.
Collapse
Affiliation(s)
- Srishti Agarwal
- Bio-Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama, Japan.
| | - Toru Maekawa
- Bio-Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama, Japan
| |
Collapse
|
21
|
Condello M, Mancini G, Meschini S. The Exploitation of Liposomes in the Inhibition of Autophagy to Defeat Drug Resistance. Front Pharmacol 2020; 11:787. [PMID: 32547395 PMCID: PMC7272661 DOI: 10.3389/fphar.2020.00787] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/12/2020] [Indexed: 12/24/2022] Open
Abstract
Autophagy is a mechanism involved in many human diseases and in cancers can have a cytotoxic/cytostatic or protective action, being in the latter case involved in multidrug resistance. Understanding which of these roles autophagy has in cancer is thus fundamental for therapeutical decisions because it permits to optimize the therapeutical approach by activating or inhibiting autophagy according to the progression of the disease. However, a serious drawback of cancer treatment is often the scarce availability of drugs and autophagy modulators at the sites of interest. In the recent years, several nanocarriers have been developed and investigated to improve the solubility, bioavailability, controlled release of therapeutics and increase their cytotoxic effect on cancer cell. Here we have reviewed only liposomes as carriers of chemotherapeutics and autophagy inhibitors because they have low toxicity and immunogenicity and they are biodegradable and versatile. In this review after the analysis of the dual role of autophagy, of the main autophagic pathways, and of the role of autophagy in multidrug resistance, we will focus on the most effective liposomal formulations, thus highlighting the great potential of these targeting systems to defeat cancer diseases.
Collapse
Affiliation(s)
- Maria Condello
- National Center for Drug Research and Evaluation, National Institute of Health, Rome, Italy
| | - Giovanna Mancini
- Institute for Biological Systems, National Research Council, Rome, Italy
| | - Stefania Meschini
- National Center for Drug Research and Evaluation, National Institute of Health, Rome, Italy
| |
Collapse
|
22
|
Chen Q, Zheng W, Zhu L, Yao D, Wang C, Song Y, Hu S, Liu H, Bai Y, Pan Y, Zhang J, Guan J, Shao C. ANXA6 Contributes to Radioresistance by Promoting Autophagy via Inhibiting the PI3K/AKT/mTOR Signaling Pathway in Nasopharyngeal Carcinoma. Front Cell Dev Biol 2020; 8:232. [PMID: 32373608 PMCID: PMC7176914 DOI: 10.3389/fcell.2020.00232] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 03/19/2020] [Indexed: 12/14/2022] Open
Abstract
Radiotherapy is a conventional and effective treatment method for nasopharyngeal carcinoma (NPC), although it can fail, mainly because radioresistance results in residual or recurrent tumors. However, the mechanisms and predictive markers of NPC radioresistance are still obscure. In this study, we identified Annexin A6 (ANXA6) as a candidate radioresistance marker by using Tandem Mass Tag quantitative proteomic analysis of NPC cells and gene chip analysis of NPC clinical samples with different radiosensitivities. It was observed that a high expression level of ANXA6 was positively correlated with radioresistance of NPC and that inhibition of ANXA6 by siRNA increased the radiosensitivity. The incidence of autophagy was enhanced in the established radioresistant NPC cells in comparison with their parent cells, and silencing autophagy with LC3 siRNA (siLC3) sensitized NPC cells to irradiation. Furthermore, ANXA6 siRNA (siANXA6) suppressed cellular autophagy by activating the PI3K/AKT/mTOR pathway, ultimately leading to radiosensitization. The combination of siANXA6 and CAL101 (an inhibitor of PI3K, p-AKT, and mTOR, concurrently) significantly reversed the above siANAX6-reduced autophagy. Suppression of PI3K/AKT/mTOR by CAL101 also increased the expression of ANXA6 in a negative feedback process. In conclusion, this study revealed for the first time that ANXA6 could promote autophagy by inhibiting the PI3K/AKT/mTOR pathway and that it thus contributes to radioresistance of NPC. The significance of this is that ANXA6 could be applied as a new predictive biomarker of NPC prognosis after radiotherapy.
Collapse
Affiliation(s)
- Qianping Chen
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wang Zheng
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lin Zhu
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dan Yao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chen Wang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yimeng Song
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Songling Hu
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongxia Liu
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yang Bai
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Pan
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianghong Zhang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Guan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chunlin Shao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
23
|
Xu Z, Han X, Ou D, Liu T, Li Z, Jiang G, Liu J, Zhang J. Targeting PI3K/AKT/mTOR-mediated autophagy for tumor therapy. Appl Microbiol Biotechnol 2019; 104:575-587. [PMID: 31832711 DOI: 10.1007/s00253-019-10257-8] [Citation(s) in RCA: 406] [Impact Index Per Article: 67.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/05/2019] [Accepted: 11/12/2019] [Indexed: 12/11/2022]
Abstract
Autophagy is a highly conserved catabolic process and participates in a variety of cellular biological activities. The phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway, as a critical regulator of autophagy, is involved in the initiation and promotion of a series of pathological disorders including various tumors. Autophagy also participates in regulating the balance between the tumor and the tumor microenvironment. Natural products have been considered a treasure of new drug discoveries and are of great value to medicine. Mounting evidence has suggested that numerous natural products are targeting PI3K/AKT/mTOR-mediated autophagy, thereby suppressing tumor growth. Furthermore, autophagy plays a "double-edged sword" role in different tumors. Targeting PI3K/AKT/mTOR-mediated autophagy is an important therapeutic strategy for a variety of tumors, and plays important roles in enhancing the chemosensitivity of tumor cells and avoiding drug resistance. Therefore, we summarized the roles of PI3K/AKT/mTOR-mediated autophagy in tumorigenesis, progression, and drug resistance of tumors, which may be utilized to design preferably therapeutic strategies for various tumors.
Collapse
Affiliation(s)
- Zhenru Xu
- Department of Rheumatology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Xu Han
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Daming Ou
- Department of Rheumatology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Ting Liu
- Department of Rheumatology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Zunxiong Li
- University of South China, Hengyang, Hunan, China
| | - Guanmin Jiang
- Department of Clinical Laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Jing Liu
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.
| | - Ji Zhang
- Department of Rheumatology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China.
| |
Collapse
|
24
|
Qu F, Wang P, Zhang K, Shi Y, Li Y, Li C, Lu J, Liu Q, Wang X. Manipulation of Mitophagy by "All-in-One" nanosensitizer augments sonodynamic glioma therapy. Autophagy 2019; 16:1413-1435. [PMID: 31674265 PMCID: PMC7480814 DOI: 10.1080/15548627.2019.1687210] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Limited penetration of chemotherapeutic drugs through the blood brain barrier (BBB), and the increased chemo-resistance of glioma cells due to macroautophagy/autophagy, result in high tumor recurrence and extremely limited survival of glioma patients. Ultrasound-targeted microbubble destruction (UTMD) is a technique of transient and reversible BBB disruption, which greatly facilitates intracerebral drug delivery. In addition, sonodynamic therapy (SDT) based on ultrasound stimulation and a sonosensitizer, can be a safe and noninvasive strategy for treating glioma. We innovatively designed a smart "all-in-one" nanosensitizer platform by incorporating the sonoactive chlorin e6 (Ce6) and an autophagy inhibitor-hydroxychloroquine (HCQ) into angiopep-2 peptide-modified liposomes (designated as ACHL), which integrates multiple diagnostic and therapeutic functions. ACHL selectively accumulated in the brain tumors during the optimal time-window of transient UTMD-mediated BBB opening. The nanosensitizer then responded to a second ultrasonic stimulation, and simultaneously unloaded HCQ and generated ROS in the glioma cells. The sonotherapy triggered apoptosis as well as MAPK/p38-PINK1-PRKN-dependent mitophagy, in which the antioxidant relieved the sonotoxicity and MAPK/p38 activation, while the inhibition of MAPK/p38 attenuated the progression toward mitophagy by compromising redistribution of PRKN. Moreover, HCQ blocking autophagosome degradation, augmented intracellular ROS production and resulted in an oxidative-damage regenerative loop. ACHL-SDT treatment using this construct significantly inhibited the xenograft-tumor growth and prolonged the survival time of tumor-bearing mice, exhibiting an improved therapeutic efficiency. All together, we demonstrated a precision sonotherapy with simultaneous apoptosis induction and mitophagy inhibition, which served as an intelligently strategic sense of working alongside, providing new insights into the theranostics of brain tumors. ABBREVIATIONS ACHL: Angiopep-2-modified liposomes loaded with Ce6 and hydroxychloroquine; ACL: Angiopep-2-modified liposomes loaded with Ce6; BBB: blood brain barrier; Ce6: chlorin e6; CHL: liposomes loaded with Ce6 and hydroxychloroquine; CL: liposomes loaded with Ce6; CNS: central nervous system; DDS: drug delivery system; EB: Evans blue; FUS: focused ultrasound; HCQ: hydroxychloroquine; LRP1: low density lipoprotein receptor-related protein 1; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MAPK: mitogen-activated protein kinase; MBs: microbubbles; MTG: MitoTracker Green; MTR: MitoTracker Red; MTT: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; PBS: phosphate-buffered saline; PDI: polydispersity index; PINK1: PTEN induced kinase 1; PRKN/parkin: parkin RBR E3 ubiquitin protein ligase; ROS: reactive oxygen species; SDT: sonodynamic therapy; SQSTM1: sequestome 1; TA: terephthalic acid; TEM: transmission electron microscopy; TUNEL: terminal deoxynucleotidyl transferase mediated dUTP nick-end labeling; US: ultrasound; UTMD: ultrasound-targeted microbubble destruction.
Collapse
Affiliation(s)
- Fei Qu
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education; College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Pan Wang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education; College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Kun Zhang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education; College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yin Shi
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education; College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yixiang Li
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education; College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Chengren Li
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Army Medical University, Chongqing, China
| | - Junhan Lu
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education; College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Quanhong Liu
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education; College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xiaobing Wang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education; College of Life Sciences, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
25
|
Howarth A, Madureira PA, Lockwood G, Storer LCD, Grundy R, Rahman R, Pilkington GJ, Hill R. Modulating autophagy as a therapeutic strategy for the treatment of paediatric high-grade glioma. Brain Pathol 2019; 29:707-725. [PMID: 31012506 PMCID: PMC8028648 DOI: 10.1111/bpa.12729] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 04/17/2019] [Indexed: 12/18/2022] Open
Abstract
Paediatric high-grade gliomas (pHGG) represent a therapeutically challenging group of tumors. Despite decades of research, there has been minimal improvement in treatment and the clinical prognosis remains poor. Autophagy, a highly conserved process for recycling metabolic substrates is upregulated in pHGG, promoting tumor progression and evading cell death. There is significant crosstalk between autophagy and a plethora of critical cellular pathways, many of which are dysregulated in pHGG. The following article will discuss our current understanding of autophagy signaling in pHGG and the potential modulation of this network as a therapeutic target.
Collapse
Affiliation(s)
- Alison Howarth
- Brain Tumour Research Centre, Institute of Biomedical and Biomolecular Sciences, IBBSUniversity of PortsmouthPortsmouthUK
| | - Patricia A. Madureira
- Brain Tumour Research Centre, Institute of Biomedical and Biomolecular Sciences, IBBSUniversity of PortsmouthPortsmouthUK
- Centre for Biomedical Research (CBMR)University of AlgarveFaroPortugal
| | - George Lockwood
- Children’s Brain Tumour Research Centre, School of Medicine, Queen’s Medical CentreUniversity of NottinghamNottinghamUK
| | - Lisa C. D. Storer
- Children’s Brain Tumour Research Centre, School of Medicine, Queen’s Medical CentreUniversity of NottinghamNottinghamUK
| | - Richard Grundy
- Children’s Brain Tumour Research Centre, School of Medicine, Queen’s Medical CentreUniversity of NottinghamNottinghamUK
| | - Ruman Rahman
- Children’s Brain Tumour Research Centre, School of Medicine, Queen’s Medical CentreUniversity of NottinghamNottinghamUK
| | - Geoffrey J. Pilkington
- Brain Tumour Research Centre, Institute of Biomedical and Biomolecular Sciences, IBBSUniversity of PortsmouthPortsmouthUK
| | - Richard Hill
- Brain Tumour Research Centre, Institute of Biomedical and Biomolecular Sciences, IBBSUniversity of PortsmouthPortsmouthUK
| |
Collapse
|
26
|
Yang K, Niu L, Bai Y, Le W. Glioblastoma: Targeting the autophagy in tumorigenesis. Brain Res Bull 2019; 153:334-340. [PMID: 31580908 DOI: 10.1016/j.brainresbull.2019.09.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 09/26/2019] [Indexed: 12/15/2022]
Abstract
Glioblastoma (GBM) is one of the most malignant and aggressive primary brain tumor, with a mean life expectancy of less than 15 months. The malignant nature of GBM prompts the need for further research on its tumorigenesis and novel treatments to improve its outcome. One of the promising research targets is autophagy, a fundamental metabolic process of degrading and recycling cellular components. Interventions to activate or inhibit autophagy have both been proposed as GBM therapies, suggesting a controversial, context-dependent role of autophagy in GBM tumorigenesis. In this review, we highlight the molecular links between GBM and autophagy with the focus on the effects of autophagy on the stemness maintenance, metabolism and proteostasis in GBM tumorigenesis. Understanding the molecular pathways involved in autophagy target is critical for GBM therapy.
Collapse
Affiliation(s)
- Kang Yang
- Department of Neurosurgery, The 2nd Hospital of Dalian Medical University, Dalian, PR China
| | - Long Niu
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, The 1st Hospital of Dalian Medical University, Dalian, PR China; Liaoning Provincial Key Laboratory for Research on Pathogenic Mechanisms of Neurological Diseases, The 1st Hospital of Dalian Medical University, Dalian, PR China
| | - Yijing Bai
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, The 1st Hospital of Dalian Medical University, Dalian, PR China; Liaoning Provincial Key Laboratory for Research on Pathogenic Mechanisms of Neurological Diseases, The 1st Hospital of Dalian Medical University, Dalian, PR China
| | - Weidong Le
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, The 1st Hospital of Dalian Medical University, Dalian, PR China; Liaoning Provincial Key Laboratory for Research on Pathogenic Mechanisms of Neurological Diseases, The 1st Hospital of Dalian Medical University, Dalian, PR China.
| |
Collapse
|
27
|
Kwon Y, Kim M, Jung HS, Kim Y, Jeoung D. Targeting Autophagy for Overcoming Resistance to Anti-EGFR Treatments. Cancers (Basel) 2019; 11:cancers11091374. [PMID: 31527477 PMCID: PMC6769649 DOI: 10.3390/cancers11091374] [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: 07/29/2019] [Revised: 08/30/2019] [Accepted: 09/10/2019] [Indexed: 12/19/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) plays critical roles in cell proliferation, tumorigenesis, and anti-cancer drug resistance. Overexpression and somatic mutations of EGFR result in enhanced cancer cell survival. Therefore, EGFR can be a target for the development of anti-cancer therapy. Patients with cancers, including non-small cell lung cancers (NSCLC), have been shown to response to EGFR-tyrosine kinase inhibitors (EGFR-TKIs) and anti-EGFR antibodies. However, resistance to these anti-EGFR treatments has developed. Autophagy has emerged as a potential mechanism involved in the acquired resistance to anti-EGFR treatments. Anti-EGFR treatments can induce autophagy and result in resistance to anti-EGFR treatments. Autophagy is a programmed catabolic process stimulated by various stimuli. It promotes cellular survival under these stress conditions. Under normal conditions, EGFR-activated phosphoinositide 3-kinase (PI3K)/AKT serine/threonine kinase (AKT)/mammalian target of rapamycin (mTOR) signaling inhibits autophagy while EGFR/rat sarcoma viral oncogene homolog (RAS)/mitogen-activated protein kinase kinase (MEK)/mitogen-activated protein kinase (MAPK) signaling promotes autophagy. Thus, targeting autophagy may overcome resistance to anti-EGFR treatments. Inhibitors targeting autophagy and EGFR signaling have been under development. In this review, we discuss crosstalk between EGFR signaling and autophagy. We also assess whether autophagy inhibition, along with anti-EGFR treatments, might represent a promising approach to overcome resistance to anti-EGFR treatments in various cancers. In addition, we discuss new developments concerning anti-autophagy therapeutics for overcoming resistance to anti-EGFR treatments in various cancers.
Collapse
Affiliation(s)
- Yoojung Kwon
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chunchon 24341, Korea.
| | - Misun Kim
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chunchon 24341, Korea.
| | - Hyun Suk Jung
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chunchon 24341, Korea.
| | - Youngmi Kim
- Institute of New Frontier Research, College of Medicine, Hallym University, Chunchon 24251, Korea.
| | - Dooil Jeoung
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chunchon 24341, Korea.
| |
Collapse
|
28
|
Liu LQ, Wang SB, Shao YF, Shi JN, Wang W, Chen WY, Ye ZQ, Jiang JY, Fang QX, Zhang GB, Xuan ZX. Hydroxychloroquine potentiates the anti-cancer effect of bevacizumab on glioblastoma via the inhibition of autophagy. Biomed Pharmacother 2019; 118:109339. [PMID: 31545270 DOI: 10.1016/j.biopha.2019.109339] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/26/2019] [Accepted: 08/05/2019] [Indexed: 12/31/2022] Open
Abstract
Bevacizumab (BEV) is widely used for the treatment of patients with recurrent glioblastoma (GBM), but recent evidence demonstrated that BEV induced cytoprotective autophagy, which allows tumor cells to survive. Hydroxychloroquine (HCQ) inhibits lysosomal acidification and blocks autophagy via influencing autophagosome fusion and degradation. HCQ is often used to enhance the efficacy of chemoradiotherapy. However, whether HCQ sensitizes GBM cells to BEV and the molecular mechanism of this effect are not clear. We showed that high concentrations of BEV increased the LC3-II/LC3-I ratio and caused the degradation of Beclin1 in the LN18 and LN229 cell lines, indicating that high concentrations of BEV induced the autophagy of the LN18 and LN229 cells. However, BEV (100 μg/ml) did not influence the autophagy of the LN18 and LN229 cells, and HCQ at less than 5 μg/ml significantly accumulated LC3B-II and p62 proteins and blocked the autophagy process. Importantly, we found that HCQ (5 μg/ml) potentiated the anti-cancer effect of BEV (100 μg/ml). Therefore, HCQ is a novel strategy that may augment the efficacy of BEV for GBM via the inhibition of autophagy.
Collapse
Affiliation(s)
- Lin-Qing Liu
- Department of Geriatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China
| | - Shi-Bing Wang
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province & Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Yan-Fei Shao
- Department of Pharmacy, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Jia-Na Shi
- Department of Pharmacy, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Wei Wang
- Department of Pharmacy, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Wan-Yuan Chen
- Department of Pathology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Zi-Qi Ye
- Department of Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Jin-Ying Jiang
- Department of Pharmacy, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Qing-Xia Fang
- Department of Pharmacy, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Guo-Bing Zhang
- Department of Pharmacy, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Zi-Xue Xuan
- Department of Pharmacy, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China.
| |
Collapse
|
29
|
Pan H, Wang Y, Na K, Wang Y, Wang L, Li Z, Guo C, Guo D, Wang X. Autophagic flux disruption contributes to Ganoderma lucidum polysaccharide-induced apoptosis in human colorectal cancer cells via MAPK/ERK activation. Cell Death Dis 2019; 10:456. [PMID: 31186406 PMCID: PMC6560101 DOI: 10.1038/s41419-019-1653-7] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/30/2019] [Accepted: 05/13/2019] [Indexed: 01/02/2023]
Abstract
Targeting autophagy may serve as a promising strategy for cancer therapy. Ganoderma lucidum polysaccharide (GLP) has been shown to exert promising anti-cancer effects. However, the underlying mechanisms remain elusive. Whether GLP regulates autophagy in cancer has never been reported. In this study, GLP induced the initiation of autophagy in colorectal cancer (CRC) HT-29 and HCT116 cells, as evidenced by enhanced level of LC3-II protein, GFP-LC3 puncta, and increased formation of double membrane vacuoles. However, GLP treatment caused marked increase of p62 expression. Addition of late stage autophagy inhibitor, chloroquine (CQ), further enhanced LC3-II and p62 level, as well as increased autophagosome accumulation, suggesting a blockage of autophagic flux by GLP in CRC cells. We then found GLP blocked autophagosome and lysosome fusion as determined by mRFP-GFP-LC3 colocalization analysis. Mechanistic study revealed that GLP-induced disruption of autophagosome-lysosome fusion is due to reduced lysosome acidification and lysosomal cathepsin activities. Cell viability and flow cytometry assays revealed that GLP-induced autophagosome accumulation is responsible for GLP-induced apoptosis in CRC cells. In line with this, inhibition of autophagy initiation by 3-methyladenine (3-MA), an early stage autophagy inhibitor, attenuated GLP-induced apoptosis. In contrast, suppression of autophagy at late stage by CQ enhanced the anti-cancer effect of GLP. Furthermore, we demonstrated that GLP-induced autophagosome accumulation and apoptosis is mediated via MAPK/ERK activation. Finally, GLP inhibited tumor growth and also inhibited autophagic flux in vivo. These results unveil new molecular mechanism underlying anti-cancer effects of GLP, suggesting that GLP is a potent autophagy inhibitor and might be useful in anticancer therapy.
Collapse
Affiliation(s)
- Haitao Pan
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China
| | - Yujie Wang
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China
| | - Kun Na
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China
| | - Ying Wang
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China
| | - Lu Wang
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China
| | - Zhenhao Li
- Zhejiang Shouxiangu Institute of Rare Medicine Plant, 12, Huanglong 3rd Road, 321200, Wuyi, Zhejiang, China
| | - Chengjie Guo
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China
| | - Dandan Guo
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China
| | - Xingya Wang
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China.
| |
Collapse
|
30
|
Wang J, Wang C, Hu X, Yu C, Zhou L, Ding Z, Zhou M. Gefitinib-mediated apoptosis is enhanced via inhibition of autophagy by chloroquine diphosphate in cutaneous squamous cell carcinoma cells. Oncol Lett 2019; 18:368-374. [PMID: 31289508 PMCID: PMC6540344 DOI: 10.3892/ol.2019.10308] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 03/21/2019] [Indexed: 12/13/2022] Open
Abstract
The development of cutaneous squamous cell carcinoma (cSCC) is associated with activation of the epidermal growth factor receptor (EGFR). EGFR-targeting presents a promising strategy for improving therapeutic efficacy. However, recent studies have suggested that tumours overexpressing EGFR depend on autophagy for survival and exhibit resistance to EGFR-targeting drugs. Chloroquine diphosphate (CQ), an autophagy inhibitor that may enhance the cytocidal effect of gefitinib against cSCC, was used in the present study. Cytotoxicity assays were performed to determine the half-maximal inhibitory concentration values of gefitinib and CQ in A431 cells. Drug interaction was analysed using CompuSyn software, which also determined combination index and dose reduction index values. Apoptosis and autophagy of A431 cells were investigated via flow cytometry, western blotting analyses, acridine orange/ethidium bromide staining and monodansylcadaverine staining. Suppression of autophagy by CQ, which was demonstrated by an alteration in microtubule associated protein 1 light chain 3-B in CQ pre-treated A431 cells, significantly enhanced cell apoptosis, which suggested that gefitinib-induced autophagy is cytoprotective. Thus, CQ was demonstrated to exhibit a synergistic apoptotic effect when used in combination with gefitinib during cSCC therapy. Further in vivo investigations are required to confirm the results of the present study.
Collapse
Affiliation(s)
- Jianyu Wang
- Department of Radiation Medicine, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Chaopeng Wang
- Department of Radiation Medicine, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xia Hu
- Department of Radiation Medicine, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Chang Yu
- Department of Radiation Medicine, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Liang Zhou
- Department of Radiation Medicine, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Zhenhua Ding
- Department of Radiation Medicine, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Meijuan Zhou
- Department of Radiation Medicine, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
31
|
Saleem H, Kulsoom Abdul U, Küçükosmanoglu A, Houweling M, Cornelissen FMG, Heiland DH, Hegi ME, Kouwenhoven MCM, Bailey D, Würdinger T, Westerman BA. The TICking clock of EGFR therapy resistance in glioblastoma: Target Independence or target Compensation. Drug Resist Updat 2019; 43:29-37. [PMID: 31054489 DOI: 10.1016/j.drup.2019.04.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 04/07/2019] [Accepted: 04/09/2019] [Indexed: 12/22/2022]
Abstract
Targeted therapy against driver mutations responsible for cancer progression has been shown to be effective in many tumor types. For glioblastoma (GBM), the epidermal growth factor receptor (EGFR) gene is the most frequently mutated oncogenic driver and has therefore been considered an attractive target for therapy. However, so far responses to EGFR-pathway inhibitors have been disappointing. We performed an exhaustive analysis of the mechanisms that might account for therapy resistance against EGFR inhibition. We define two major mechanisms of resistance and propose modalities to overcome them. The first resistance mechanism concerns target independence. In this case, cells have lost expression of the EGFR protein and experience no negative impact of EGFR targeting. Loss of extrachromosomally encoded EGFR as present in double minute DNA is a frequent mechanism for this type of drug resistance. The second mechanism concerns target compensation. In this case, cells will counteract EGFR inhibition by activation of compensatory pathways that render them independent of EGFR signaling. Compensatory pathway candidates are platelet-derived growth factor β (PDGFβ), Insulin-like growth factor 1 (IGFR1) and cMET and their downstream targets, all not commonly mutated at the time of diagnosis alongside EGFR mutation. Given that both mechanisms make cells independent of EGFR expression, other means have to be found to eradicate drug resistant cells. To this end we suggest rational strategies which include the use of multi-target therapies that hit truncation mutations (mechanism 1) or multi-target therapies to co-inhibit compensatory proteins (mechanism 2).
Collapse
Affiliation(s)
- Hamza Saleem
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Amsterdam University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands
| | - U Kulsoom Abdul
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Amsterdam University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands
| | - Asli Küçükosmanoglu
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Amsterdam University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands
| | - Megan Houweling
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Amsterdam University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands
| | - Fleur M G Cornelissen
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Amsterdam University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands; Division of Biology, Nature Science Building, 9500 Gilman Drive, CA, 92093-0377, United States
| | - Dieter H Heiland
- Department of Neurosurgery, Medical Center - University of Freiburg, Baden-Württemberg, Germany
| | - Monika E Hegi
- Department of Clinical Neurosciences, Lausanne University Hospital, Chemin des Boveresses 155, CH-1066 Epalinges, Switzerland
| | - Mathilde C M Kouwenhoven
- Department of Neurology, Brain Tumor Center Amsterdam, Amsterdam University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands
| | - David Bailey
- IOTA Pharmaceuticals Ltd, St Johns Innovation Centre, Cowley Road, Cambridge, CB4 0WS, UK
| | - Tom Würdinger
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Amsterdam University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands
| | - Bart A Westerman
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Amsterdam University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands.
| |
Collapse
|
32
|
Abstract
Resistance to therapy is one of the prime causes for treatment failure in cancer and recurrent disease. In recent years, autophagy has emerged as an important cell survival mechanism in response to different stress conditions that are associated with cancer treatment and aging. Autophagy is an evolutionary conserved catabolic process through which damaged cellular contents are degraded after uptake into autophagosomes that subsequently fuse with lysosomes for cargo degradation, thereby alleviating stress. In addition, autophagy serves to maintain cellular homeostasis by enriching nutrient pools. Although autophagy can act as a double-edged sword at the interface of cell survival and cell death, increasing evidence suggest that in the context of cancer therapy-induced stress responses, it predominantly functions as a cell survival mechanism. Here, we provide an up-to-date overview on our current knowledge of the role of pro-survival autophagy in cancer therapy at the preclinical and clinical stages and delineate the molecular mechanisms of autophagy regulation in response to therapy-related stress conditions. A better understanding of the interplay of cancer therapy and autophagy may allow to unveil new targets and avenues for an improved treatment of therapy-resistant tumors in the foreseeable future.
Collapse
|
33
|
Serrano-Saenz S, Palacios C, Delgado-Bellido D, López-Jiménez L, Garcia-Diaz A, Soto-Serrano Y, Casal JI, Bartolomé RA, Fernández-Luna JL, López-Rivas A, Oliver FJ. PIM kinases mediate resistance of glioblastoma cells to TRAIL by a p62/SQSTM1-dependent mechanism. Cell Death Dis 2019; 10:51. [PMID: 30718520 PMCID: PMC6362213 DOI: 10.1038/s41419-018-1293-3] [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: 07/03/2018] [Revised: 12/07/2018] [Accepted: 12/18/2018] [Indexed: 12/21/2022]
Abstract
Glioblastoma (GBM) is the most common and aggressive brain tumor and is associated with poor prognosis. GBM cells are frequently resistant to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and finding new combinatorial therapies to sensitize glioma cells to TRAIL remains an important challenge. PIM kinases are serine/threonine kinases that promote cell survival and proliferation and are highly expressed in different tumors. In this work, we studied the role of PIM kinases as regulators of TRAIL sensitivity in GBM cells. Remarkably, PIM inhibition or knockdown facilitated activation by TRAIL of a TRAIL-R2/DR5-mediated and mitochondria-operated apoptotic pathway in TRAIL-resistant GBM cells. The sensitizing effect of PIM knockdown on TRAIL-induced apoptosis was mediated by enhanced caspase-8 recruitment to and activation at the death-inducing signaling complex (DISC). Interestingly, TRAIL-induced internalization of TRAIL-R2/DR5 was significantly reduced in PIM knockdown cells. Phospho-proteome profiling revealed a decreased phosphorylation of p62/SQSTM1 after PIM knockdown. Our results also showed an interaction between p62/SQSTM1 and the DISC that was reverted after PIM knockdown. In line with this, p62/SQSTM1 ablation increased TRAIL-R2/DR5 levels and facilitated TRAIL-induced caspase-8 activation, revealing an inhibitory role of p62/SQSTM1 in TRAIL-mediated apoptosis in GBM. Conversely, upregulation of TRAIL-R2/DR5 upon PIM inhibition and apoptosis induced by the combination of PIM inhibitor and TRAIL were abrogated by a constitutively phosphorylated p62/SQSTM1S332E mutant. Globally, our data represent the first evidence that PIM kinases regulate TRAIL-induced apoptosis in GBM and identify a specific role of p62/SQSTM1Ser332 phosphorylation in the regulation of the extrinsic apoptosis pathway activated by TRAIL.
Collapse
Affiliation(s)
- Santiago Serrano-Saenz
- Instituto de Parasitología y Biomedicina López-Neyra, CSIC, CIBERONC, Parque Tecnológico Ciencias de la Salud, Avenida del Conocimiento, s/n, 18100, Armilla, Granada, Spain.,Centro de Investigación Biomédica en Red-Oncología (CIBERONC), Carlos III Health Institute, Madrid, Spain
| | - Carmen Palacios
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC), Carlos III Health Institute, Madrid, Spain.,Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, CIBERONC, Avda Américo Vespucio 24, 41092, Sevilla, Spain
| | - Daniel Delgado-Bellido
- Instituto de Parasitología y Biomedicina López-Neyra, CSIC, CIBERONC, Parque Tecnológico Ciencias de la Salud, Avenida del Conocimiento, s/n, 18100, Armilla, Granada, Spain
| | - Laura López-Jiménez
- Instituto de Parasitología y Biomedicina López-Neyra, CSIC, CIBERONC, Parque Tecnológico Ciencias de la Salud, Avenida del Conocimiento, s/n, 18100, Armilla, Granada, Spain
| | - Angel Garcia-Diaz
- Instituto de Parasitología y Biomedicina López-Neyra, CSIC, CIBERONC, Parque Tecnológico Ciencias de la Salud, Avenida del Conocimiento, s/n, 18100, Armilla, Granada, Spain
| | - Yolanda Soto-Serrano
- Instituto de Parasitología y Biomedicina López-Neyra, CSIC, CIBERONC, Parque Tecnológico Ciencias de la Salud, Avenida del Conocimiento, s/n, 18100, Armilla, Granada, Spain
| | - J Ignacio Casal
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28039, Madrid, Spain
| | - Rubén A Bartolomé
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28039, Madrid, Spain
| | - José Luis Fernández-Luna
- HUMV-Hospital Universitario Marqués de Valdecilla Avenida Valdecilla, 25, 39008, Santander, Cantabria, Spain
| | - Abelardo López-Rivas
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC), Carlos III Health Institute, Madrid, Spain. .,Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, CIBERONC, Avda Américo Vespucio 24, 41092, Sevilla, Spain.
| | - F Javier Oliver
- Instituto de Parasitología y Biomedicina López-Neyra, CSIC, CIBERONC, Parque Tecnológico Ciencias de la Salud, Avenida del Conocimiento, s/n, 18100, Armilla, Granada, Spain. .,Centro de Investigación Biomédica en Red-Oncología (CIBERONC), Carlos III Health Institute, Madrid, Spain.
| |
Collapse
|
34
|
Trejo-Solís C, Serrano-Garcia N, Escamilla-Ramírez Á, Castillo-Rodríguez RA, Jimenez-Farfan D, Palencia G, Calvillo M, Alvarez-Lemus MA, Flores-Nájera A, Cruz-Salgado A, Sotelo J. Autophagic and Apoptotic Pathways as Targets for Chemotherapy in Glioblastoma. Int J Mol Sci 2018; 19:ijms19123773. [PMID: 30486451 PMCID: PMC6320836 DOI: 10.3390/ijms19123773] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/14/2018] [Accepted: 11/21/2018] [Indexed: 01/07/2023] Open
Abstract
Glioblastoma multiforme is the most malignant and aggressive type of brain tumor, with a mean life expectancy of less than 15 months. This is due in part to the high resistance to apoptosis and moderate resistant to autophagic cell death in glioblastoma cells, and to the poor therapeutic response to conventional therapies. Autophagic cell death represents an alternative mechanism to overcome the resistance of glioblastoma to pro-apoptosis-related therapies. Nevertheless, apoptosis induction plays a major conceptual role in several experimental studies to develop novel therapies against brain tumors. In this review, we outline the different components of the apoptotic and autophagic pathways and explore the mechanisms of resistance to these cell death pathways in glioblastoma cells. Finally, we discuss drugs with clinical and preclinical use that interfere with the mechanisms of survival, proliferation, angiogenesis, migration, invasion, and cell death of malignant cells, favoring the induction of apoptosis and autophagy, or the inhibition of the latter leading to cell death, as well as their therapeutic potential in glioma, and examine new perspectives in this promising research field.
Collapse
Affiliation(s)
- Cristina Trejo-Solís
- Departamento de Neuroinmunología, Laboratorio de Neurobiología Molecular y Celular, Laboratorio Experimental de Enfermedades Neurodegenerativas del Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", C.P. 14269 Ciudad de México, Mexico.
| | - Norma Serrano-Garcia
- Departamento de Neuroinmunología, Laboratorio de Neurobiología Molecular y Celular, Laboratorio Experimental de Enfermedades Neurodegenerativas del Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", C.P. 14269 Ciudad de México, Mexico.
| | - Ángel Escamilla-Ramírez
- Departamento de Neuroinmunología, Laboratorio de Neurobiología Molecular y Celular, Laboratorio Experimental de Enfermedades Neurodegenerativas del Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", C.P. 14269 Ciudad de México, Mexico.
- Hospital Regional de Alta Especialidad de Oaxaca, Secretaria de Salud, C.P. 71256 Oaxaca, Mexico.
| | | | - Dolores Jimenez-Farfan
- Laboratorio de Inmunología, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, C.P. 04510 Ciudad de México, Mexico.
| | - Guadalupe Palencia
- Departamento de Neuroinmunología, Laboratorio de Neurobiología Molecular y Celular, Laboratorio Experimental de Enfermedades Neurodegenerativas del Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", C.P. 14269 Ciudad de México, Mexico.
| | - Minerva Calvillo
- Departamento de Neuroinmunología, Laboratorio de Neurobiología Molecular y Celular, Laboratorio Experimental de Enfermedades Neurodegenerativas del Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", C.P. 14269 Ciudad de México, Mexico.
| | - Mayra A Alvarez-Lemus
- División Académica de Ingeniería y Arquitectura, Universidad Juárez Autónoma de Tabasco, C.P. 86040 Tabasco, Mexico.
| | - Athenea Flores-Nájera
- Departamento de Cirugía Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Secretaria de Salud, 14000 Ciudad de México, Mexico.
| | - Arturo Cruz-Salgado
- Departamento de Neuroinmunología, Laboratorio de Neurobiología Molecular y Celular, Laboratorio Experimental de Enfermedades Neurodegenerativas del Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", C.P. 14269 Ciudad de México, Mexico.
| | - Julio Sotelo
- Departamento de Neuroinmunología, Laboratorio de Neurobiología Molecular y Celular, Laboratorio Experimental de Enfermedades Neurodegenerativas del Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", C.P. 14269 Ciudad de México, Mexico.
| |
Collapse
|
35
|
Chen F, Song Q, Yu Q. Axl inhibitor R428 induces apoptosis of cancer cells by blocking lysosomal acidification and recycling independent of Axl inhibition. Am J Cancer Res 2018; 8:1466-1482. [PMID: 30210917 PMCID: PMC6129480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 07/09/2018] [Indexed: 06/08/2023] Open
Abstract
R428 (BGB324) is an anti-cancer drug candidate under clinical investigation. It inhibits the receptor tyrosine kinase Axl and induces apoptosis of many types of cancer cells, but the relationship between the two has not been well established. We investigated the molecular mechanisms of the R428-induced apoptosis and found that R428 induced extensive cytoplasmic vacuolization and caspase activation, independent of its inhibitory effects on Axl. Further analyses revealed that R428 blocked lysosomal acidification and recycling, accumulated autophagosomes and lysosomes, and induced cell apoptosis. Inhibition of autophagy by autophagy inhibitors or autophagic gene-knockout alleviated the R428-induced vacuoles formation and cell apoptosis. Our study uncovered a novel function and mechanism of R428 in addition to its ability to inhibit Axl. These data will help to better direct the application of R428 as an anti-cancer reagent. It also adds new knowledge to understand the regulation of autophagy and apoptosis.
Collapse
Affiliation(s)
- Fangfang Chen
- Shanghai Institute of Materia Medica, Chinese Academy of SciencesShanghai 201203, China
- University of Chinese Academy of SciencesBeijing 100049, China
| | - Qiaoling Song
- Shanghai Institute of Materia Medica, Chinese Academy of SciencesShanghai 201203, China
- University of Chinese Academy of SciencesBeijing 100049, China
| | - Qiang Yu
- Shanghai Institute of Materia Medica, Chinese Academy of SciencesShanghai 201203, China
- University of Chinese Academy of SciencesBeijing 100049, China
| |
Collapse
|
36
|
Zhang YJ, Xu ZG, Li SQ, He LJ, Tang Y, Chen ZZ, Yang DL. Benzimidazoisoquinoline derivatives inhibit glioblastoma cell proliferation through down-regulating Raf/MEK/ERK and PI3K/AKT pathways. Cancer Cell Int 2018; 18:90. [PMID: 29988358 PMCID: PMC6022716 DOI: 10.1186/s12935-018-0588-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 06/14/2018] [Indexed: 01/09/2023] Open
Abstract
Background Recent studies showed that benzimidazoleisoquinolinone derivatives exhibit anticancer activity against human cancer cell lines. The aim of this study is to evaluate the anti-tumor effects and mechanisms of benzimidazoleisoquinolinones in isocitrate dehydrogenase-wildtype subtype of human glioblastoma (GBM) cells. Methods Human U87 and LN229 cell lines were used to perform the experiments. MTT was applied to screen the effective small molecular inhibitors suppressing growth of GBM cells. Colony formation and BrdU staining assays were performed to assess the inhibition effect of compound-1H on the proliferation of GBM cells. The cell cycle and apoptosis were measured by flow cytometry and western blot to analyze the changes of the relative protein expressions and their signal pathways. Results Compound-1H could suppress GBM cells in a time- and dose-dependent manner. Treatment of compound-1H could arrest cell cycle in S phase through up-regulating P21 and P53, and down-regulating cyclin A and E in a dose-dependent manner. Compound-1H also induced mitochondrial-dependent apoptosis by increasing Bax, cleaved caspase-3, cleaved caspase-9 and poly ADP-ribose polymerase expression, and decreasing Bcl-2 expression. Moreover, phosphorylated (p)-AKT and p-ERK levels relating to cell proliferation were dramatically decreased in U87 and LN229 cells. Conclusions Our results suggest that it is the first time to report the compound-1H with benzimidazoleisoquinolinone core playing antitumor activity in human glioblastoma cells by inhibiting Raf/MEK/ERK and PI3K/AKT signaling pathways, and it could be as a lead compound for the further development of targeted glioblastoma cancer therapy.
Collapse
Affiliation(s)
- Ya-Jun Zhang
- Chongqing Engineering Laboratory of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, International Academy of Targeted Therapeutics and Innovation (IATTI), Chongqing University of Arts and Sciences, Chongqing, 402160 China
| | - Zhi-Gang Xu
- Chongqing Engineering Laboratory of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, International Academy of Targeted Therapeutics and Innovation (IATTI), Chongqing University of Arts and Sciences, Chongqing, 402160 China
| | - Shi-Qiang Li
- Chongqing Engineering Laboratory of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, International Academy of Targeted Therapeutics and Innovation (IATTI), Chongqing University of Arts and Sciences, Chongqing, 402160 China
| | - Liu-Jun He
- Chongqing Engineering Laboratory of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, International Academy of Targeted Therapeutics and Innovation (IATTI), Chongqing University of Arts and Sciences, Chongqing, 402160 China
| | - Yan Tang
- Chongqing Engineering Laboratory of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, International Academy of Targeted Therapeutics and Innovation (IATTI), Chongqing University of Arts and Sciences, Chongqing, 402160 China
| | - Zhong-Zhu Chen
- Chongqing Engineering Laboratory of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, International Academy of Targeted Therapeutics and Innovation (IATTI), Chongqing University of Arts and Sciences, Chongqing, 402160 China
| | - Dong-Lin Yang
- Chongqing Engineering Laboratory of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, International Academy of Targeted Therapeutics and Innovation (IATTI), Chongqing University of Arts and Sciences, Chongqing, 402160 China
| |
Collapse
|
37
|
Autophagy in glioma cells: An identity crisis with a clinical perspective. Cancer Lett 2018; 428:139-146. [PMID: 29709703 DOI: 10.1016/j.canlet.2018.04.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/14/2018] [Accepted: 04/20/2018] [Indexed: 01/06/2023]
Abstract
Over the last decade, autophagy has emerged as one of the critical cellular systems that control homeostasis. Besides management of normal homeostatic processes, autophagy can also be induced by tissue damage stress or by rapidly progressing tumors. During tumor progression, autophagy mediates a cellular reaction to the changes inside and outside of cells, which leads to tumor adaptation. Even though the regulation of autophagy seems universal and is a well-described process, its dysregulation and role in glioma progression remain an important topic of investigation. In this review, we summarize recent evidence of autophagy regulation in brain tumor tissues and possible interconnection between signaling pathways that govern cellular responses. This perspective may help to assess the qualitative differences and various outcomes in response to autophagy stimulation.
Collapse
|
38
|
Hombach-Klonisch S, Mehrpour M, Shojaei S, Harlos C, Pitz M, Hamai A, Siemianowicz K, Likus W, Wiechec E, Toyota BD, Hoshyar R, Seyfoori A, Sepehri Z, Ande SR, Khadem F, Akbari M, Gorman AM, Samali A, Klonisch T, Ghavami S. Glioblastoma and chemoresistance to alkylating agents: Involvement of apoptosis, autophagy, and unfolded protein response. Pharmacol Ther 2018; 184:13-41. [DOI: 10.1016/j.pharmthera.2017.10.017] [Citation(s) in RCA: 206] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
39
|
Xie FJ, Zheng QQ, Qin J, Zhang LL, Han N, Mao WM. Autophagy Inhibition Stimulates Apoptosis in Oesophageal Squamous Cell Carcinoma Treated with Fasudil. J Cancer 2018; 9:1050-1056. [PMID: 29581784 PMCID: PMC5868172 DOI: 10.7150/jca.23388] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 01/28/2018] [Indexed: 01/19/2023] Open
Abstract
Fasudil has been proven to be a promising chemotherapeutic drug for various malignancies. However, the potential anticancer effects of fasudil in oesophageal squamous cell carcinoma (ESCC) remain to be established. We confirmed the RhoA activity is inhibited by fasudil in ESCC cells. Then measured the effects of fasudil on apoptosis and autophagy in ESCC. Our study showed fasudil could both induce ESCCs apoptosis and autophagy, and when fasudil-induced autophagy was inhibited by knockdown of the essential autophagy genes (Beclin 1 or ATG7), and pharmacologic agent (chloroquine) treatment, both treatments also significantly sensitized ESCC to fasudil-induced apoptosis, reducing cell viability in vitro. Our study showed autophagy inhibitors combined with fasudil could significantly induce ESCC apoptosis, which may provide a novel therapeutic strategy for ESCC.
Collapse
Affiliation(s)
- Fa-Jun Xie
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou 310022, China
- Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Oesophagus), Hangzhou 310022, China
| | - Qiu-Qing Zheng
- Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Oesophagus), Hangzhou 310022, China
- Department of Ultrasonography, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Jing Qin
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou 310022, China
- Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Oesophagus), Hangzhou 310022, China
| | - Ling-Ling Zhang
- Department of Medical Oncology, Peking University International Hospital, Beijing 102206, China
| | - Na Han
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou 310022, China
- Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Oesophagus), Hangzhou 310022, China
| | - Wei-Min Mao
- Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Oesophagus), Hangzhou 310022, China
- Department of Thoracic Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, China
| |
Collapse
|
40
|
Antunes F, Pereira GJ, Jasiulionis MG, Bincoletto C, Smaili SS. Nutritional shortage augments cisplatin-effects on murine melanoma cells. Chem Biol Interact 2017; 281:89-97. [PMID: 29273566 DOI: 10.1016/j.cbi.2017.12.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/25/2017] [Accepted: 12/18/2017] [Indexed: 01/06/2023]
Abstract
Melanoma incidence increases every year worldwide and is responsible for 80% of skin cancer deaths. Due to its metastatic potential and resistance to almost any treatments such as chemo, radio, immune and targeted-therapy, the patients still have a poor prognosis, especially at metastatic stage. Considering that, it is crucial to find new therapeutic approaches to overcome melanoma resistance. Here we investigated the effect of cisplatin (CDDP), one of the chemotherapeutic agents used for melanoma treatment, in association with nutritional deprivation in murine melanoma cell lines. Cell death and autophagy were evaluated after the treatment with cisplatin, nutritional deprivation and its association using an in vitro model of murine melanocytes malignant transformation to metastatic melanoma. Our results showed that nutritional deprivation augmented cell death induced by cisplatin in melanoma cells, especially at the metastatic subtype, with slight effects on melanocytes. Mechanistic studies revealed that although autophagy was present at high levels in basal conditions in melanoma cells, was not essential for cell death process that involved mitochondrial damage, reactive oxygen species production and possible glycolysis inhibition. In conclusion, nutritional shortage in combination with chemotherapeutic drugs as cisplatin can be a valuable new therapeutic strategy to overcome melanoma resistance.
Collapse
Affiliation(s)
- F Antunes
- Universidade Federal de São Paulo, Escola Paulista de Medicina Department of Pharmacology (EPM/UNIFESP), São Paulo, SP, Brazil
| | - G J Pereira
- Universidade Federal de São Paulo, Escola Paulista de Medicina Department of Pharmacology (EPM/UNIFESP), São Paulo, SP, Brazil.
| | - M G Jasiulionis
- Universidade Federal de São Paulo, Escola Paulista de Medicina Department of Pharmacology (EPM/UNIFESP), São Paulo, SP, Brazil
| | - C Bincoletto
- Universidade Federal de São Paulo, Escola Paulista de Medicina Department of Pharmacology (EPM/UNIFESP), São Paulo, SP, Brazil
| | - S S Smaili
- Universidade Federal de São Paulo, Escola Paulista de Medicina Department of Pharmacology (EPM/UNIFESP), São Paulo, SP, Brazil.
| |
Collapse
|
41
|
Verbaanderd C, Maes H, Schaaf MB, Sukhatme VP, Pantziarka P, Sukhatme V, Agostinis P, Bouche G. Repurposing Drugs in Oncology (ReDO)-chloroquine and hydroxychloroquine as anti-cancer agents. Ecancermedicalscience 2017; 11:781. [PMID: 29225688 PMCID: PMC5718030 DOI: 10.3332/ecancer.2017.781] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Indexed: 12/26/2022] Open
Abstract
Chloroquine (CQ) and hydroxychloroquine (HCQ) are well-known 4-aminoquinoline antimalarial agents. Scientific evidence also supports the use of CQ and HCQ in the treatment of cancer. Overall, preclinical studies support CQ and HCQ use in anti-cancer therapy, especially in combination with conventional anti-cancer treatments since they are able to sensitise tumour cells to a variety of drugs, potentiating the therapeutic activity. Thus far, clinical results are mostly in favour of the repurposing of CQ. However, over 30 clinical studies are still evaluating the activity of both CQ and HCQ in different cancer types and in combination with various standard treatments. Interestingly, CQ and HCQ exert effects both on cancer cells and on the tumour microenvironment. In addition to inhibition of the autophagic flux, which is the most studied anti-cancer effect of CQ and HCQ, these drugs affect the Toll-like receptor 9, p53 and CXCR4-CXCL12 pathway in cancer cells. In the tumour stroma, CQ was shown to affect the tumour vasculature, cancer-associated fibroblasts and the immune system. The evidence reviewed in this paper indicates that both CQ and HCQ deserve further clinical investigations in several cancer types. Special attention about the drug (CQ versus HCQ), the dose and the schedule of administration should be taken in the design of new trials.
Collapse
Affiliation(s)
- Ciska Verbaanderd
- Anticancer Fund, Brussels, 1853 Strombeek-Bever, Belgium.,Cell Death Research and Therapy Lab, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium.,Clinical Pharmacology and Pharmacotherapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Hannelore Maes
- Cell Death Research and Therapy Lab, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Marco B Schaaf
- Cell Death Research and Therapy Lab, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Vikas P Sukhatme
- GlobalCures, Inc, Newton, MA 02459, USA.,Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA; Current address: Emory School of Medicine, Atlanta, GA 30322, USA
| | - Pan Pantziarka
- Anticancer Fund, Brussels, 1853 Strombeek-Bever, Belgium.,The George Pantziarka TP53 Trust, London KT1 2JP, UK
| | | | - Patrizia Agostinis
- Cell Death Research and Therapy Lab, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | | |
Collapse
|
42
|
Retinoic acid regulates endometriotic stromal cell growth through upregulation of Beclin1. Arch Gynecol Obstet 2017; 297:93-99. [PMID: 29063947 DOI: 10.1007/s00404-017-4549-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 09/20/2017] [Indexed: 10/18/2022]
Abstract
PURPOSE To elucidate the role of retinoic acid (RA) in autophagy-mediated endometriosis. METHODS The mRNA and protein expressions of autophagy markers were examined in Ishikawa cells and endometriotic stromal cells (ESCs) after RA treatment. Beclin1 expression was specifically analyzed in clinical samples of endometriosis. The effect of Beclin1 knockdown on ESC growth was assessed, and the effect of autophagy inhibition on the sensitivity of endometriotic cells to RA was analyzed. RESULTS RA treatment enhanced the autophagy in ESCs, and Beclin1 expression showed a negative correlation with the clinical stage of endometriosis. Beclin1 knockdown enhanced ESC growth, whereas RA treatment reversed this effect. Furthermore, inhibition of autophagy by chloroquine (CQ) and Beclin1 knockdown did not show any positive effect on the sensitivity of endometriotic cells to RA. CONCLUSIONS RA treatment induces autophagy and Beclin1 may play an important role in endometriosis progression.
Collapse
|
43
|
Mainz L, Rosenfeldt MT. Autophagy and cancer - insights from mouse models. FEBS J 2017; 285:792-808. [PMID: 28921866 DOI: 10.1111/febs.14274] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 08/10/2017] [Accepted: 09/13/2017] [Indexed: 01/09/2023]
Abstract
(Macro-)autophagy is an evolutionary conserved 'self-digestion program' that serves to maintain cellular metabolism and is implicated in many pathological processes such as cancer. In recent years, an increasing number of studies in murine cancer models have provided a plethora of sometimes conflicting results about the role of autophagy in cancer biology. This review summarizes these studies and raises awareness that there are situations in which autophagy blockage might indeed reduce tumor growth, but that sometimes the exact opposite is the case. It is therefore vital to mimic patient conditions in preclinical mouse experiments as thoroughly as possible before commencing clinical trials.
Collapse
Affiliation(s)
- Laura Mainz
- Institute for Pathology, Germany & Comprehensive Cancer Center Mainfranken, University of Wuerzburg, Germany
| | - Mathias T Rosenfeldt
- Institute for Pathology, Germany & Comprehensive Cancer Center Mainfranken, University of Wuerzburg, Germany
| |
Collapse
|
44
|
Wang X, Qiu Y, Yu Q, Li H, Chen X, Li M, Long Y, Liu Y, Lu L, Tang J, Zhang Z, He Q. Enhanced glioma therapy by synergistic inhibition of autophagy and tyrosine kinase activity. Int J Pharm 2017; 536:1-10. [PMID: 28887220 DOI: 10.1016/j.ijpharm.2017.09.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/19/2017] [Accepted: 09/03/2017] [Indexed: 12/19/2022]
Abstract
Autophagy is a lysosomal degradation pathway that acts as a cytoprotective mechanism causing treatment resistance in various cancer cells. Recent studies showed that hydroxychloroquine can inhibit the latter step of autophagy and therefore enhance the anti-glioma efficiency of ZD6474, a tyrosine kinase inhibitor. However, the nonselective distribution of ZD6474 in vivo and the low penetrating ability of hydroxychloroquine when crossing the blood-brain barrier restrict their clinical use in glioma therapy. Here we coencapsulated ZD6474 and hydroxychloroquine into R6dGR peptide-modified liposomes (R6dGR-Lip) which can specifically recognize both integrin αvβ3 and neuropilin-1 receptors that are highly expressed on the endothelial cells and glioma cells. R6dGR significantly enhanced the brain targeting and overcame the blood-brain barrier. Our results confirmed that loading hydroxychloroquine into R6dGR-Lip blocked autophagic flux more efficiently than free hydroxychloroquine in glioma cells and significantly sensitized glioma cells to ZD6474-induced cell death in vitro and in vivo. The coencapsulated R6dGR-modified liposomes (ZD6474/HCQ-R6dGR-Lip) prolonged the medium survival time of intracranial C6 glioma bearing mice by 1.2-fold compared with ZD6474-R6dGR-Lip, 1.5-fold compared with free ZD6474/HCQ, and 1.8-fold compared with free ZD6474, exhibiting a synergistic therapeutic effect. Therefore, ZD6474/HCQ-R6dGR-Lip is presented as a potential strategy which could be further used for efficient anti-glioma therapy.
Collapse
Affiliation(s)
- Xuhui Wang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Yue Qiu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Qianwen Yu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Hui Li
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Xiaoxiao Chen
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Man Li
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Yang Long
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Yayuan Liu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Libao Lu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Jiajing Tang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Qin He
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China.
| |
Collapse
|
45
|
Li H, You L, Xie J, Pan H, Han W. The roles of subcellularly located EGFR in autophagy. Cell Signal 2017; 35:223-230. [PMID: 28428083 DOI: 10.1016/j.cellsig.2017.04.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/14/2017] [Accepted: 04/15/2017] [Indexed: 12/11/2022]
Abstract
The epidermal growth factor receptor (EGFR) is a well-studied receptor-tyrosine kinase that serves vital roles in regulation of organ development and cancer progression. EGFR not only exists on the plasma membrane, but also widely expressed in the nucleus, endosomes, and mitochondria. Most recently, several lines of evidences indicated that autophagy is regulated by EGFR in kinase-active and -independent manners. In this review, we summarized recent advances in our understanding of the functions of different subcellularly located EGFR on autophagy. Specifically, plasma membrane- and cytoplasm-located EGFR (pcEGFR) acts as a tyrosine kinase to regulate autophagy via the PI3K/AKT1/mTOR, RAS/MAPK1/3, and STAT3 signaling pathways. The kinase-independent function of pcEGFR inhibits autophagy by maintaining SLC5A1-regulated intracellular glucose level. Endosome-located EGFR phosphorylates and inhibits Beclin1 to suppress autophagy, while kinase-independent endosome-located EGFR releases Beclin1 from the Rubicon-Beclin1 complex to increase autophagy. Additionally, the nuclear EGFR activates PRKDC/PNPase/MYC signaling to inhibit autophagy. Although the role of mitochondria-located EGFR in autophagy is largely unexplored, the production of ATP and reactive oxygen species mediated by mitochondrial dynamics is most likely to influence autophagy.
Collapse
Affiliation(s)
- Hongsen Li
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Liangkun You
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiansheng Xie
- Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
46
|
Lysosomal dysfunction and autophagy blockade contribute to IMB-6G-induced apoptosis in pancreatic cancer cells. Sci Rep 2017; 7:41862. [PMID: 28139733 PMCID: PMC5282566 DOI: 10.1038/srep41862] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 01/04/2017] [Indexed: 01/08/2023] Open
Abstract
Targeting the autophagic pathway is currently regarded as an attractive strategy for cancer drug discovery. Our previous work showed that IMB-6G is a novel N-substituted sophoridinic acid derivative with potent cytotoxicity against tumor cells, yet the effect of IMB-6G on autophagy and pancreatic cancer cell death remains unknown. Here, we show that IMB-6G inhibits the growth of MiaPaCa-2 and HupT-3 pancreatic cancer cells and induces caspase-mediated apoptosis, which is correlated with an accumulation of autophagic vacuoles. IMB-6G promotes autophagosome accumulation from the early stage of treatment but blocks autophagic flux in the degradation stage, mainly through attenuation of lysosomal cathepsin activity in pancreatic cancer cells. Moreover, IMB-6G triggers lysosomal membrane permeabilization (LMP), followed by cathepsin B/CTSB and cathepsin D/CTSD release from lysosomes into the cytoplasm. Inhibition of autophagosome formation with siRNA against autophagy protein 5 (Atg5) attenuates IMB-6G-induced LMP and apoptosis. Furthermore, cathepsin inhibitors relieve IMB-6G-induced apoptosis as well. Altogether, our findings demonstrate that IMB-6G is a novel autophagy inhibitor, which induces autophagy-dependent apoptosis through autophagosomal-cathepsin axis in pancreatic cancer cells and indicate the potential value of IMB-6G as a novel antitumor drug candidate.
Collapse
|
47
|
Glioblastoma, hypoxia and autophagy: a survival-prone 'ménage-à-trois'. Cell Death Dis 2016; 7:e2434. [PMID: 27787518 PMCID: PMC5133985 DOI: 10.1038/cddis.2016.318] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 08/24/2016] [Accepted: 09/09/2016] [Indexed: 12/14/2022]
Abstract
Glioblastoma multiforme is the most common and the most aggressive primary brain tumor. It is characterized by a high degree of hypoxia and also by a remarkable resistance to therapy because of its adaptation capabilities that include autophagy. This degradation process allows the recycling of cellular components, leading to the formation of metabolic precursors and production of adenosine triphosphate. Hypoxia can induce autophagy through the activation of several autophagy-related proteins such as BNIP3, AMPK, REDD1, PML, and the unfolded protein response-related transcription factors ATF4 and CHOP. This review summarizes the most recent data about induction of autophagy under hypoxic condition and the role of autophagy in glioblastoma.
Collapse
|
48
|
Detection of autophagy in Hirschsprung's disease: implication for its role in aganglionosis. Neuroreport 2016; 26:1044-50. [PMID: 26509546 DOI: 10.1097/wnr.0000000000000465] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Hirschsprung's disease (HD) is a common congenital gastrointestinal malformation, characterized by the lack of ganglion cells from the distal rectum to the proximal bowel, but the pathogenesis is not well understood. This paper evaluates the effects of autophagy in HD. Using electron microscopy, the autophagosomes were detected in three segments: narrow segment (NS), transitional segment (TS), and dilated segment (DS). Typical autophagosome structures are found in the Auerbach plexus of both NS and TS. Real-time PCR results showed that Beclin1 (NS vs. TS, P<0.01) and LC3 (NS vs. TS, P<0.05) mRNA were the highest in the NS, but p75 (NS vs. TS, P<0.01) was the highest in the DS. Correlation analysis results showed a positive correlation between Beclin1 and LC3 mRNA levels (R=0.736, P=0.000), whereas inverse correlations were found between p75 and Beclin1/LC3 mRNA levels (p75 vs. Beclin1: R=-0.714, P=0.000; p75 vs. LC3: R=-0.619, P=0.000). Immunohistochemistry analyses indicated a consistent result with mRNA levels, by increased Beclin1-positive and LC3-positive neurons, but reduced p75-positive neurons in the Auerbach plexus of TS compared with DS. These findings indicated that autophagy exists in the bowel of patients with HD. On the basis of the detection of the highest expression of the autophagy genes in NS, autophagy may additionally cause the lack of neurons.
Collapse
|
49
|
Liu Y, Zheng J, Zhang Y, Wang Z, Yang Y, Bai M, Dai Y. Fucoxanthin Activates Apoptosis via Inhibition of PI3K/Akt/mTOR Pathway and Suppresses Invasion and Migration by Restriction of p38-MMP-2/9 Pathway in Human Glioblastoma Cells. Neurochem Res 2016; 41:2728-2751. [DOI: 10.1007/s11064-016-1989-7] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 04/15/2016] [Accepted: 06/22/2016] [Indexed: 10/21/2022]
|
50
|
Yang C, Wu C, Xu D, Wang M, Xia Q. AstragalosideII inhibits autophagic flux and enhance chemosensitivity of cisplatin in human cancer cells. Biomed Pharmacother 2016; 81:166-175. [DOI: 10.1016/j.biopha.2016.03.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 03/21/2016] [Indexed: 10/21/2022] Open
|