1
|
Chen Y, Yi H, Liao S, He J, Zhou Y, Lei Y. LC3B: A microtubule-associated protein influences disease progression and prognosis. Cytokine Growth Factor Rev 2025; 81:16-26. [PMID: 39701849 DOI: 10.1016/j.cytogfr.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 12/21/2024]
Abstract
Microtubule-associated protein 1 light chain 3B (MAP1LC3B, also known as LC3B) is a mammalian homolog of the autophagy-related protein 8 (ATG8) family. It plays a crucial role in cellular autophagy and is involved in several vital biological processes, including apoptosis and differentiation. Additionally, LC3B regulates immune responses. Due to its close association with malignant tumors and neurodegenerative diseases, and its potential as a prognostic indicator and therapeutic target, LC3B has become a significant research focus. This article aims to provide a comprehensive and systematic understanding of LC3B's role and mechanisms in autophagy, its impact on apoptosis and the underlying mechanisms, its regulation of cellular differentiation and transdifferentiation, its modulation of immune and inflammatory responses, the influence of upstream regulatory factors on LC3B's function, and its relevance to disease diagnosis, treatment, and prognosis. The goal is to establish a solid foundation for understanding LC3B's role in cellular processes and its regulatory mechanisms.
Collapse
Affiliation(s)
- Yan Chen
- Department of Blood Transfusion, The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University/Hunan Cancer Hospital, Changsha, Hunan 410013, China; Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China
| | - Hong Yi
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Shan Liao
- Department of Pathology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Junyu He
- Department of Clinical Laboratory, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, Hunan 410007, China
| | - Yanhong Zhou
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China.
| | - Yan Lei
- Department of Blood Transfusion, The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University/Hunan Cancer Hospital, Changsha, Hunan 410013, China.
| |
Collapse
|
2
|
Molavand M, Ebrahimnezhade N, Kiani A, Yousefi B, Nazari A, Majidinia M. Regulation of autophagy by non-coding RNAs in human glioblastoma. Med Oncol 2024; 41:260. [PMID: 39375229 DOI: 10.1007/s12032-024-02513-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/18/2024] [Indexed: 10/09/2024]
Abstract
Glioblastoma, a lethal form of brain cancer, poses substantial challenges in treatment due to its aggressive nature and resistance to standard therapies like radiation and chemotherapy. Autophagy has a crucial role in glioblastoma progression by supporting cellular homeostasis and promoting survival under stressful conditions. Non-coding RNAs (ncRNAs) play diverse biological roles including, gene regulation, chromatin remodeling, and the maintenance of cellular homeostasis. Emerging evidence reveals the intricate regulatory mechanisms of autophagy orchestrated by non-coding RNAs (ncRNAs) in glioblastoma. The diverse roles of these ncRNAs in regulating crucial autophagy-related pathways, including AMPK/mTOR signaling, the PI3K/AKT pathway, Beclin1, and other autophagy-triggering system regulation, sheds light on ncRNAs biological mechanisms in the proliferation, invasion, and therapy response of glioblastoma cells. Furthermore, the clinical implications of targeting ncRNA-regulated autophagy as a promising therapeutic strategy for glioblastoma treatment are in the spotlight of ongoing studies. In this review, we delve into our current understanding of how ncRNAs regulate autophagy in glioblastoma, with a specific focus on microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), and their intricate interplay with therapy response.
Collapse
Affiliation(s)
- Mehran Molavand
- Student Research Commitee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Niloufar Ebrahimnezhade
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Arash Kiani
- Student Research Commite, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Bahman Yousefi
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran.
- Molecular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ahmad Nazari
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran.
- Tehran University of Medical Sciences, Tehran, Iran.
| | - Maryam Majidinia
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
3
|
Kundu M, Das S, Dey A, Mandal M. Dual perspective on autophagy in glioma: Detangling the dichotomous mechanisms of signaling pathways for therapeutic insights. Biochim Biophys Acta Rev Cancer 2024; 1879:189168. [PMID: 39121913 DOI: 10.1016/j.bbcan.2024.189168] [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: 04/16/2024] [Revised: 06/25/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Autophagy is a normal physiological process that aids the recycling of cellular nutrients, assisting the cells to cope with stressed conditions. However, autophagy's effect on cancer, including glioma, is uncertain and involves complicated molecular mechanisms. Several contradictory reports indicate that autophagy may promote or suppress glioma growth and progression. Autophagy inhibitors potentiate the efficacy of chemotherapy or radiation therapy in glioma. Numerous compounds stimulate autophagy to cause glioma cell death. Autophagy is also involved in the therapeutic resistance of glioma. This review article aims to detangle the complicated molecular mechanism of autophagy to provide a better perception of the two-sided role of autophagy in glioma and its therapeutic implications. The protein and epigenetic modulators of the cytoprotective and cytotoxic role of autophagy are described in this article. Moreover, several signaling pathways are associated with autophagy and its effects on glioma. We have reviewed the molecular pathways and highlighted the signaling axis involved in cytoprotective and cytotoxic autophagy. Additionally, this article discusses the role of autophagy in therapeutic resistance, including glioma stem cell maintenance and tumor microenvironment regulation. It also summarizes several investigations on the anti-glioma effects of autophagy modulators to understand the associated mechanisms and provide insights regarding its therapeutic implications.
Collapse
Affiliation(s)
- Moumita Kundu
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India; Center for Multidisciplinary Research & Innovations, Brainware University, Barasat, India; Department of Pharmaceutical Technology, Brainware University, Barasat, India.
| | - Subhayan Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India; Department of Allied Health Sciences, Brainware University, Barasat, India
| | - Ankita Dey
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| |
Collapse
|
4
|
Kusaczuk M, Ambel ET, Naumowicz M, Velasco G. Cellular stress responses as modulators of drug cytotoxicity in pharmacotherapy of glioblastoma. Biochim Biophys Acta Rev Cancer 2024; 1879:189054. [PMID: 38103622 DOI: 10.1016/j.bbcan.2023.189054] [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: 07/28/2023] [Revised: 11/21/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023]
Abstract
Despite the extensive efforts to find effective therapeutic strategies, glioblastoma (GBM) remains a therapeutic challenge with dismal prognosis of survival. Over the last decade the role of stress responses in GBM therapy has gained a great deal of attention, since depending on the duration and intensity of these cellular programs they can be cytoprotective or promote cancer cell death. As such, initiation of the UPR, autophagy or oxidative stress may either impede or facilitate drug-mediated cell killing. In this review, we summarize the mechanisms that regulate ER stress, autophagy, and oxidative stress during GBM development and progression to later discuss the involvement of these stress pathways in the response to different treatments. We also discuss how a precise understanding of the molecular mechanisms regulating stress responses evoked by different pharmacological agents could decisively contribute to the design of novel and more effective combinational treatments against brain malignancies.
Collapse
Affiliation(s)
- Magdalena Kusaczuk
- Department of Pharmaceutical Biochemistry, Medical University of Bialystok, Mickiewicza 2A, 15-222 Bialystok, Poland.
| | - Elena Tovar Ambel
- Department of Biochemistry and Molecular Biology, School of Biology, Complutense University, Instituto de Investigación Sanitaria San Carlos IdISSC, 28040 Madrid, Spain
| | - Monika Naumowicz
- Department of Physical Chemistry, Faculty of Chemistry, University of Bialystok, K. Ciolkowskiego 1K, 15-245 Bialystok, Poland
| | - Guillermo Velasco
- Department of Biochemistry and Molecular Biology, School of Biology, Complutense University, Instituto de Investigación Sanitaria San Carlos IdISSC, 28040 Madrid, Spain.
| |
Collapse
|
5
|
Won J, Lee S, Ahmad Khan Z, Choi J, Ho Lee T, Hong Y. Suppression of DAPK1 reduces ischemic brain injury through inhibiting cell death signaling and promoting neural remodeling. Brain Res 2023; 1820:148588. [PMID: 37742938 DOI: 10.1016/j.brainres.2023.148588] [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: 05/17/2023] [Revised: 08/11/2023] [Accepted: 09/10/2023] [Indexed: 09/26/2023]
Abstract
The role of death-associated protein kinase1 (DAPK1) in post-stroke functional recovery is controversial, as is its mechanism of action and any neural remodeling effect after ischemia. To assess the debatable role of DAPK1, we established the middle cerebral artery occlusion (MCAo) model in DAPK1 knockout mice and Sprague-Dawley (SD) rats. We identified that the genetic deletion of the DAPK1 as well as pharmacological inhibition of DAPK1 showed reduced brain infarct volume and neurological deficit. We report that DAPK1 inhibition (DI) reduces post-stroke neuronal death by inhibiting BAX/BCL2 and LC3/Beclin1 mediated apoptosis and autophagy, respectively. Histological analysis displayed a reduction in nuclear condensation, neuronal dissociation, and degraded cytoplasm in the DI group. The DI treatment showed enhanced dendrite spine density and neurite outgrowth, upregulated neural proliferation marker proteins like brain-derived neurotrophic factor, and reduced structural abnormalities of the cortical pyramidal neurons. This research shows that DAPK1 drives cell death, its activation exacerbates functional recovery after cerebral ischemia and shows that oxazolone-based DI could be an excellent candidate for stroke and ischemic injury intervention.
Collapse
Affiliation(s)
- Jinyoung Won
- Department of Rehabilitation Science, Graduate School of Inje University, Gimhae, South Korea; Biohealth Products Research Center (BPRC), Inje University, Gimhae, South Korea; Research Center for Aged-life Redesign (RCAR), Inje University, Gimhae, South Korea
| | - Seunghoon Lee
- Biohealth Products Research Center (BPRC), Inje University, Gimhae, South Korea; Research Center for Aged-life Redesign (RCAR), Inje University, Gimhae, South Korea; Department of Physical Therapy, College of Healthcare Medical Science & Engineering, Inje University, Gimhae, South Korea
| | - Zeeshan Ahmad Khan
- Biohealth Products Research Center (BPRC), Inje University, Gimhae, South Korea; Research Center for Aged-life Redesign (RCAR), Inje University, Gimhae, South Korea; Department of Physical Therapy, College of Healthcare Medical Science & Engineering, Inje University, Gimhae, South Korea
| | - Jeonghyun Choi
- Department of Rehabilitation Science, Graduate School of Inje University, Gimhae, South Korea; Biohealth Products Research Center (BPRC), Inje University, Gimhae, South Korea; Research Center for Aged-life Redesign (RCAR), Inje University, Gimhae, South Korea
| | - Tae Ho Lee
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Science, Fujian Medical University, Fuzhou, China
| | - Yonggeun Hong
- Department of Rehabilitation Science, Graduate School of Inje University, Gimhae, South Korea; Biohealth Products Research Center (BPRC), Inje University, Gimhae, South Korea; Research Center for Aged-life Redesign (RCAR), Inje University, Gimhae, South Korea; Department of Physical Therapy, College of Healthcare Medical Science & Engineering, Inje University, Gimhae, South Korea.
| |
Collapse
|
6
|
Chen J, Rodriguez AS, Morales MA, Fang X. Autophagy Modulation and Its Implications on Glioblastoma Treatment. Curr Issues Mol Biol 2023; 45:8687-8703. [PMID: 37998723 PMCID: PMC10670099 DOI: 10.3390/cimb45110546] [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: 09/26/2023] [Revised: 10/26/2023] [Accepted: 10/26/2023] [Indexed: 11/25/2023] Open
Abstract
Autophagy is a vital cellular process that functions to degrade and recycle damaged organelles into basic metabolites. This allows a cell to adapt to a diverse range of challenging conditions. Autophagy assists in maintaining homeostasis, and it is tightly regulated by the cell. The disruption of autophagy has been associated with many diseases, such as neurodegenerative disorders and cancer. This review will center its discussion on providing an in-depth analysis of the current molecular understanding of autophagy and its relevance to brain tumors. We will delve into the current literature regarding the role of autophagy in glioma pathogenesis by exploring the major pathways of JAK2/STAT3 and PI3K/AKT/mTOR and summarizing the current therapeutic interventions and strategies for glioma treatment. These treatments will be evaluated on their potential for autophagy induction and the challenges associated with their utilization. By understanding the mechanism of autophagy, clinical applications for future therapeutics in treating gliomas can be better targeted.
Collapse
Affiliation(s)
- Johnny Chen
- Department of Neuroscience, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA;
| | - Andrea Salinas Rodriguez
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA;
| | - Maximiliano Arath Morales
- Department of Biology, College of Science, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA;
| | - Xiaoqian Fang
- Department of Neuroscience, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA;
| |
Collapse
|
7
|
Shen N, Wang L, Wu J, Chen X, Hu F, Su Y. Meta‑analysis of the autophagy‑associated protein LC3 as a prognostic marker in colorectal cancer. Exp Ther Med 2023; 26:492. [PMID: 37753301 PMCID: PMC10518644 DOI: 10.3892/etm.2023.12191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 08/02/2023] [Indexed: 09/28/2023] Open
Abstract
Microtubule-associated protein 1 light chain 3 (LC3) is an autophagy-associated gene, which is involved in the progression of a number of human malignancies. Such as Breast Cancer, Liver Cancer, and Lung Cancer. However, the role of LC3 in colorectal cancer (CC) remains to be fully elucidated. Therefore, the prognostic role of LC3 expression in CC was evaluated in the present study, with an emphasis on the clinicopathology and prognosis. Expression of LC3 in CC was examined using PubMed, Cochrane Library, Excerpta Medica Database, China Knowledge Infrastructure and Wanfang Data. Newcastle-Ottawa scale was used to screen the literature quality, and RevMan 5.4 and STATA 14.0 were used for the meta-analysis. A total of 1,689 patients from 10 studies were included in the present meta-analysis. The findings of the present study suggested that increased LC3 expression levels were associated with histological grade [odds ratio (OR)=0.91, 95% confidence interval (CI) (0.47, 1.77), P<0.001] and TNM stage [OR=0.91, 95% CI (0.47, 1.77), P<0.001], but were not associated with sex [OR=1.14, 95% CI (0.90, 1.51)], age [OR=0.89, 95% CI (0.67, 1.20)], tumor size [OR=0.78, 95% CI (0.30, 2.34)], histological grade [OR=0.82, 95% CI (0.43, 1.95)] and lymph node metastasis [OR=2.05, 95% CI (1.19, 3.60)] in CC. In addition, the increased expression of LC3 was revealed to be a prognostic factor for the overall survival of patients with CC. In conclusion, the autophagy-associated protein LC3 may be a prognostic indicator of human CC.
Collapse
Affiliation(s)
- Ning Shen
- Department of Oncology, Zhangqiu People's Hospital, Jinan, Shandong 250200, P.R. China
| | - Lijuan Wang
- Blood Purification Center, Suzhou Science and Technology Town Hospital, Suzhou, Jiangsu 215153, P.R. China
| | - Jingjing Wu
- Department of Oncology, Zhangqiu People's Hospital, Jinan, Shandong 250200, P.R. China
| | - Xuefang Chen
- Blood Purification Center, Suzhou Science and Technology Town Hospital, Suzhou, Jiangsu 215153, P.R. China
| | - Fengchao Hu
- Department of Oncology, Zhangqiu People's Hospital, Jinan, Shandong 250200, P.R. China
| | - Yi Su
- Quality Management Office, Zhangqiu People's Hospital, Jinan, Shandong 250200, P.R. China
| |
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
|
Chen T, Zeng C, Li Z, Wang J, Sun F, Huang J, Lu S, Zhu J, Zhang Y, Sun X, Zhen Z. Investigation of chemoresistance to first-line chemotherapy and its possible association with autophagy in high-risk neuroblastoma. Front Oncol 2022; 12:1019106. [PMID: 36338726 PMCID: PMC9632338 DOI: 10.3389/fonc.2022.1019106] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 10/05/2022] [Indexed: 11/28/2022] Open
Abstract
High-risk neuroblastoma (NB) is sensitive to chemotherapy but susceptible to chemoresistance. In this study, we aimed to analyze the incidence of chemoresistance in high-risk NB patients and to explore the role of autophagy in NB chemoresistance. We retrospectively analyzed the incidence of changing the chemotherapy regimen due to disease stabilization or disease progression during induction chemotherapy in high-risk NB patients, which was expressed as the chemoresistance rate. The autophagy levels were probed in tumor cells exposed to first-line chemotherapy agents. The sensitivity of tumor cells to chemotherapy agents and apoptosis rate were observed after inhibiting autophagy by transfection of shRNA or chloroquine (CQ). This study included 247 patients with high-risk NB. The chemoresistance rates of patients treated with cyclophosphamide + adriamycin + vincristine (CAV) alternating with etoposide + cisplatin (EP) (Group 1) and CAV alternating with etoposide + ifosfamide + cisplatin (VIP) (Group 2) was 61.5% and 39.9% (P = 0.0009), respectively. Group 2 had better survival rates than group 1. After exposure to cisplatin, cyclophosphamide, and etoposide, the autophagy-related proteins LC3-I, LC3-II, and Beclin-1 were upregulated, and the incidence of autophagy vesicle formation and the expression of P62 were increased. Chemotherapeutic agents combined with CQ significantly increased the chemotherapeutic sensitivity of tumor cells and increased the cell apoptosis. The downregulated expression of Beclin-1 increased the sensitivity of tumor cells to chemotherapeutics. Our results suggest that increasing the chemotherapy intensity can overcome resistance to NB. Inhibition of autophagy is beneficial to increase the sensitivity of NB to chemotherapy agents.
Collapse
Affiliation(s)
- Tingting Chen
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Chenggong Zeng
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zhuoran Li
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Juan Wang
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Feifei Sun
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Junting Huang
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Suying Lu
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jia Zhu
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yizhuo Zhang
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xiaofei Sun
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zijun Zhen
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- *Correspondence: Zijun Zhen,
| |
Collapse
|
10
|
Protein Quality Control in Glioblastoma: A Review of the Current Literature with New Perspectives on Therapeutic Targets. Int J Mol Sci 2022; 23:ijms23179734. [PMID: 36077131 PMCID: PMC9456419 DOI: 10.3390/ijms23179734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/12/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
Protein quality control allows eukaryotes to maintain proteostasis under the stress of constantly changing conditions. In this review, we discuss the current literature on PQC, highlighting flaws that must exist for malignancy to occur. At the nidus of PQC, the expression of BAG1-6 reflects the cell environment; each isoform directs proteins toward different, parallel branches of the quality control cascade. The sum of these branches creates a net shift toward either homeostasis or apoptosis. With an established role in ALP, Bag3 is necessary for cell survival in stress conditions including those of the cancerous niche (i.e., hypoxia, hypermutation). Evidence suggests that excessive Bag3–HSP70 activity not only sustains, but also propagates cancers. Its role is anti-apoptotic—which allows malignant cells to persist—and intercellular—with the production of infectious ‘oncosomes’ enabling cancer expansion and recurrence. While Bag3 has been identified as a key prognostic indicator in several cancer types, its investigation is limited regarding glioblastoma. The cochaperone HSP70 has been strongly linked with GBM, while ALP inhibitors have been shown to improve GBM susceptibility to chemotherapeutics. Given the highly resilient, frequently recurrent nature of GBM, the targeting of Bag3 is a necessary consideration for the successful and definitive treatment of GBM.
Collapse
|
11
|
Shi J, Yang N, Han M, Qiu C. Emerging roles of ferroptosis in glioma. Front Oncol 2022; 12:993316. [PMID: 36072803 PMCID: PMC9441765 DOI: 10.3389/fonc.2022.993316] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 07/28/2022] [Indexed: 11/27/2022] Open
Abstract
Glioma is the most common primary malignant tumor in the central nervous system, and directly affects the quality of life and cognitive function of patients. Ferroptosis, is a new form of regulated cell death characterized by iron-dependent lipid peroxidation. Ferroptosis is mainly due to redox imbalance and involves multiple intracellular biology processes, such as iron metabolism, lipid metabolism, and antioxidants synthesis. Induction of ferroptosis could be a new target for glioma treatment, and ferroptosis-related processes are associated with chemoresistance and radioresistance in glioma. In the present review, we provide the characteristics, key regulators and pathways of ferroptosis and the crosstalk between ferroptosis and other programmed cell death in glioma, we also proposed the application and prospect of ferroptosis in the treatment of glioma.
Collapse
Affiliation(s)
- Jiaqi Shi
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Ning Yang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- Department of Epidemiology and Health Statistics, School of Public Health, Shandong University, Jinan, China
| | - Mingzhi Han
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chen Qiu
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Chen Qiu,
| |
Collapse
|
12
|
Improper Proteostasis: Can It Serve as Biomarkers for Neurodegenerative Diseases? Mol Neurobiol 2022; 59:3382-3401. [PMID: 35305242 DOI: 10.1007/s12035-022-02775-w] [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: 10/22/2021] [Accepted: 02/19/2022] [Indexed: 10/18/2022]
Abstract
Cells synthesize new proteins after multiple molecular decisions. Damage of existing proteins, accumulation of abnormal proteins, and basic requirement of new proteins trigger protein quality control (PQC)-based alternative strategies to cope against proteostasis imbalance. Accumulation of misfolded proteins is linked with various neurodegenerative disorders. However, how deregulated components of this quality control system and their lack of general mechanism-based long-term changes can serve as biomarkers for neurodegeneration remains largely unexplored. Here, our article summarizes the chief findings, which may facilitate the search of novel and relevant proteostasis mechanism-based biomarkers associated with neuronal disorders. Understanding the abnormalities of PQC coupled molecules as possible biomarkers can help to determine neuronal fate and their contribution to the aetiology of several nervous system disorders.
Collapse
|
13
|
Fu X, Hong L, Gong H, Kan G, Zhang P, Cui TT, Fan G, Si X, Zhu J. Identification of a Nomogram with an Autophagy-Related Risk Signature for Survival Prediction in Patients with Glioma. Int J Gen Med 2022; 15:1517-1535. [PMID: 35210825 PMCID: PMC8857975 DOI: 10.2147/ijgm.s335571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
Background Glioma is a common type of tumor in the central nervous system characterized by high morbidity and mortality. Autophagy plays vital roles in the development and progression of glioma, and is involved in both normal physiological and various pathophysiological progresses. Patients and Methods A total of 531 autophagy-related genes (ARGs) were obtained and 1738 glioma patients were collected from three public databases. We performed least absolute shrinkage and selection operator regression to identify the optimal prognosis-related genes and constructed an autophagy-related risk signature. The performance of the signature was validated by receiver operating characteristic analysis, survival analysis, clinic correlation analysis, and Cox regression. A nomogram model was established by using multivariate Cox regression analysis. Schoenfeld’s global and individual test were used to estimate time-varying covariance for the assumption of the Cox proportional hazard regression analysis. The R programming language was used as the main data analysis and visualizing tool. Results An overall survival-related risk signature consisting of 15 ARGs was constructed and significantly stratified glioma patients into high- and low-risk groups (P < 0.0001). The area under the ROC curve of 1-, 3-, 5-year survival was 0.890, 0.923, and 0.889, respectively. Univariate and multivariate Cox analyses indicated that the risk signature was a satisfactory independent prognostic factor. Moreover, a nomogram model integrating risk signature with clinical information for predicting survival rates of patients with glioma was constructed (C-index=0.861±0.024). Conclusion This study constructed a novel and reliable ARG-related risk signature, which was verified as a satisfactory prognostic marker. The nomogram model could provide a reference for individually predicting the prognosis for each patient with glioma and promoting the selection of optimal treatment.
Collapse
Affiliation(s)
- Xiaofeng Fu
- Department of Ultrasound, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310000, People’s Republic of China
| | - Luwei Hong
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310000, People’s Republic of China
| | - Haiying Gong
- Department of Ultrasound, Yiwu Traditional Chinese Medicine Hospital, Jinhua, Zhejiang, 321000, People’s Republic of China
| | - Guangjuan Kan
- Department of Ultrasound, The Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310000, People’s Republic of China
| | - Pengfei Zhang
- Department of Ultrasound, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310000, People’s Republic of China
| | - Ting-Ting Cui
- Department of Ultrasound, Taizhou Traditional Chinese Medicine Hospital, Taizhou, Zhejiang, 318000, People’s Republic of China
| | - Gonglin Fan
- Department of Ultrasound, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310000, People’s Republic of China
| | - Xing Si
- Hangzhou Normal University, Hangzhou, Zhejiang, 310000, People’s Republic of China
| | - Jiang Zhu
- Department of Ultrasound, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310000, People’s Republic of China
- Correspondence: Jiang Zhu, Department of Ultrasound, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 31000, People’s Republic of China, Tel +86 13757122629, Email
| |
Collapse
|
14
|
Belyaeva E, Kharwar RK, Ulasov IV, Karlina I, Timashev P, Mohammadinejad R, Acharya A. Isoforms of autophagy-related proteins: role in glioma progression and therapy resistance. Mol Cell Biochem 2022; 477:593-604. [PMID: 34854022 DOI: 10.1007/s11010-021-04308-w] [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: 08/04/2021] [Accepted: 11/19/2021] [Indexed: 11/26/2022]
Abstract
Autophagy is the process of recycling and utilization of degraded organelles and macromolecules in the cell compartments formed during the fusion of autophagosomes with lysosomes. During autophagy induction the healthy and tumor cells adapt themselves to harsh conditions such as cellular stress or insufficient supply of nutrients in the cell environment to maintain their homeostasis. Autophagy is currently seen as a form of programmed cell death along with apoptosis and necroptosis. In recent years multiple studies have considered the autophagy as a potential mechanism of anticancer therapy in malignant glioma. Although, subsequent steps in autophagy development are known and well-described, on molecular level the mechanism of autophagosome initiation and maturation using autophagy-related proteins is under investigation. This article reviews current state about the mechanism of autophagy, its molecular pathways and the most recent studies on roles of autophagy-related proteins and their isoforms in glioma progression and its treatment.
Collapse
Affiliation(s)
- Elizaveta Belyaeva
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, Russia, 119991
| | - Rajesh Kumar Kharwar
- Endocrine Research Laboratory, Department of Zoology, Kutir Post Graduate College, Chakkey, Jaunpur, UP, India
| | - Ilya V Ulasov
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, Russia, 119991.
| | - Irina Karlina
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, Russia, 119991
| | - Petr Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation, 119991
- Department of Polymers and Composites, N.N.Semenov Institute of Chemical Physics, 4 Kosygin st., Moscow, Russian Federation, 119991
- Chemistry Department, Lomonosov Moscow State University, Leninskiye Gory 1-3, Moscow, Russian Federation, 119991
| | - Reza Mohammadinejad
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Arbind Acharya
- Tumor Immunology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, UP, 221005, India.
| |
Collapse
|
15
|
Li Q, Xia L, Sun C, Zhang H, Zheng M, Zhang H, Lu H, Wang Z. Role of Borneol Induced Autophagy in Enhancing Radiosensitivity of Malignant Glioma. Front Oncol 2021; 11:749987. [PMID: 34917504 PMCID: PMC8668811 DOI: 10.3389/fonc.2021.749987] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/01/2021] [Indexed: 11/24/2022] Open
Abstract
Glioma is the common primary craniocerebral malignancy with unfavorable prognosis. It is currently treated by surgical resection supplemented by radiotherapy, although the resistance of glioma cells to radiation limits the therapeutic outcomes. The aim of the present study was to determine the potential radiosensitizing effects of borneol and the underlying mechanisms. We found that borneol administration along with radiotherapy significantly inhibited the growth of primary glioma cells in vitro and in vivo. Furthermore, borneol markedly increased the number of autophagosomes in the glioma cells, which coincided with increased expression of beclin-1 and LC3. And the combination of borneol and radiation exposure significantly decreased the expression levels of HIF-1α, mTORC1 and eIF4E. In addition, silencing mTORC1 and eIF4E upregulated Beclin-1 and LC3 and decreased the expression of HIF-1α, thereby inhibiting tumor cell proliferation. Our findings suggest that borneol sensitizes glioma cells to radiation by inducing autophagy via inhibition of the mTORC1/eIF4E/HIF-1α regulatory axis.
Collapse
Affiliation(s)
- Qinglin Li
- Department of Scientific Research, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Liang Xia
- Department of Neurotumor Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Caixing Sun
- Department of Neurotumor Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Huangjie Zhang
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Mengying Zheng
- Department of Medical School, Zhejiang University City College, Hangzhou, China
| | - Hongyan Zhang
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Hongyang Lu
- Department of Thoracic Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Zeng Wang
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
16
|
Abstract
Around three out of one hundred thousand people are diagnosed with glioblastoma multiforme, simply called glioblastoma, which is the most common primary brain tumor in adults. With a dismal prognosis of a little over a year, receiving a glioblastoma diagnosis is oftentimes fatal. A major advancement in its treatment was made almost two decades ago when the alkylating chemotherapeutic agent temozolomide (TMZ) was combined with radiotherapy (RT). Little progress has been made since then. Therapies that focus on the modulation of autophagy, a key process that regulates cellular homeostasis, have been developed to curb the progression of glioblastoma. The dual role of autophagy (cell survival or cell death) in glioblastoma has led to the development of autophagy inhibitors and promoters that either work as monotherapies or as part of a combination therapy to induce cell death, cellular senescence, and counteract the ability of glioblastoma stem cells (GSCs) for initiating tumor recurrence. The myriad of cellular pathways that act upon the modulation of autophagy have created contention between two groups: those who use autophagy inhibition versus those who use promotion of autophagy to control glioblastoma growth. We discuss rationale for using current major therapeutics, their molecular mechanisms for modulation of autophagy in glioblastoma and GSCs, their potentials for making strides in combating glioblastoma progression, and their possible shortcomings. These shortcomings may fuel the innovation of novel delivery systems and therapies involving TMZ in conjunction with another agent to pave the way towards a new gold standard of glioblastoma treatment.
Collapse
Affiliation(s)
- Amanda J Manea
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, 6439 Garners Ferry Road, Columbia, SC, 29209, USA
| | - Swapan K Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, 6439 Garners Ferry Road, Columbia, SC, 29209, USA.
| |
Collapse
|
17
|
Establishment and validation of five autophagy-related signatures for predicting survival and immune microenvironment in glioma. Genes Genomics 2021; 44:79-95. [PMID: 34609723 DOI: 10.1007/s13258-021-01172-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 09/23/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Gliomas, especially Glioblastoma multiforme, are the most frequent type of primary tumors in central nervous system. Increasing researches have revealed the relationship between autophagy and tumor, while the molecular mechanism of autophagy in glioma is still rarely reported. OBJECTIVE Our research aims to conform the autophagy-related genes (ARGs) implicated in the development and progression of glioma and improve our understanding of autophagy in glioma. METHODS 20 candidate ARGs were screened through the protein-protein interaction network. We also downloaded the publicly accessible glioma data for 665 individuals from TCGA and 970 individuals from CGGA with RNA sequences and clinicopathological information. Subsequently, univariate and multivariate Cox regression analysis identified 5 key ARGs among the 20 candidate genes as key prognostic genes for survival, GSEA and immune response analysis. RESULTS ATG5, BCL2L1, CASP3, CASP8, GAPDH were identified as key ARGs in our research. Further studies showed that the high-risk population was linked to a dismal prognosis and suggested an immune-inhibitory microenvironment. GSEA results demonstrated that high risk population was closely related to DNA repair, hypoxia pathways, implicated in immunosuppression and carcinogenesis. Through CMap, we finally identified 14 candidate drugs for the ARG high risk population. CONCLUSIONS This study established and verified an ARG risk model, which can serve as an independent predictor for prognosis, reflect on the strength of the immune response and predict the potential drugs in glioma. Our findings offer new understandings of ARG molecular mechanism and promising therapeutic targets for glioma treatment.
Collapse
|
18
|
Pelaz SG, Ollauri-Ibáñez C, Lillo C, Tabernero A. Impairment of Autophagic Flux Participates in the Antitumor Effects of TAT-Cx43 266-283 in Glioblastoma Stem Cells. Cancers (Basel) 2021; 13:cancers13174262. [PMID: 34503072 PMCID: PMC8428230 DOI: 10.3390/cancers13174262] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/13/2021] [Accepted: 08/21/2021] [Indexed: 11/20/2022] Open
Abstract
Simple Summary Autophagy is a process in which the cell recycles components that are not needed at that moment and uses the resulting elements to satisfy more urgent needs. Depending on the specific context, this can be beneficial or detrimental for tumor development. We found that in glioblastoma, the most lethal brain tumor, autophagy is upregulated and contributes to glioblastoma stem cell survival under starvation. Importantly, the antitumor peptide TAT-Cx43266-283 blocks autophagy flux, contributing to the death of glioblastoma stem cells. This peptide induces glioblastoma stem cell death in nutrient-deprived and complete environments, while the effect of other unsuccessful drugs for glioblastoma depends on nutrient context, supporting the potential of TAT-Cx43266-283 as a treatment to improve the lives of glioblastoma patients. Abstract Autophagy is a physiological process by which various damaged or non-essential cytosolic components are recycled, contributing to cell survival under stress conditions. In cancer, autophagy can have antitumor or protumor effects depending on the developmental stage. Here, we use Western blotting, immunochemistry, and transmission electron microscopy to demonstrate that the antitumor peptide TAT-Cx43266-283, a c-Src inhibitor, blocks autophagic flux in glioblastoma stem cells (GSCs) under basal and nutrient-deprived conditions. Upon nutrient deprivation, GSCs acquired a dormant-like phenotype that was disrupted by inhibition of autophagy with TAT-Cx43266-283 or chloroquine (a classic autophagy inhibitor), leading to GSC death. Remarkably, dasatinib, a clinically available c-Src inhibitor, could not replicate TAT-Cx43266-283 effect on dormant GSCs, revealing for the first time the possible involvement of pathways other than c-Src in TAT-Cx43266-283 effect. TAT-Cx43266-283 exerts an antitumor effect both in nutrient-complete and nutrient-deprived environments, which constitutes an advantage over chloroquine and dasatinib, whose effects depend on nutrient environment. Finally, our analysis of the levels of autophagy-related proteins in healthy and glioma donors suggests that autophagy is upregulated in glioblastoma, further supporting the interest in inhibiting this process in the most aggressive brain tumor and the potential use of TAT-Cx43266-283 as a therapy for this type of cancer.
Collapse
Affiliation(s)
- Sara G. Pelaz
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Calle Pintor Fernando Gallego 1, 37007 Salamanca, Spain; (S.G.P.); (C.O.-I.); (C.L.)
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Virgen de la Vega, 10ª Planta, Paseo de San Vicente 58-182, 37007 Salamanca, Spain
| | - Claudia Ollauri-Ibáñez
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Calle Pintor Fernando Gallego 1, 37007 Salamanca, Spain; (S.G.P.); (C.O.-I.); (C.L.)
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Virgen de la Vega, 10ª Planta, Paseo de San Vicente 58-182, 37007 Salamanca, Spain
| | - Concepción Lillo
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Calle Pintor Fernando Gallego 1, 37007 Salamanca, Spain; (S.G.P.); (C.O.-I.); (C.L.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Virgen de la Vega, 10ª Planta, Paseo de San Vicente 58-182, 37007 Salamanca, Spain
- Departamento de Biología Celular y Patología, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| | - Arantxa Tabernero
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Calle Pintor Fernando Gallego 1, 37007 Salamanca, Spain; (S.G.P.); (C.O.-I.); (C.L.)
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Virgen de la Vega, 10ª Planta, Paseo de San Vicente 58-182, 37007 Salamanca, Spain
- Correspondence:
| |
Collapse
|
19
|
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
|
20
|
Fu X, Chen S, Wang X, Shen Y, Zeng R, Wu Q, Lu Y, Shi J, Zhou S. Dendrobium nobile Lindl. alkaloids alleviate Mn-induced neurotoxicity via PINK1/Parkin-mediated mitophagy in PC12 cells. Biochem Biophys Rep 2021; 26:100877. [PMID: 33889759 PMCID: PMC8047462 DOI: 10.1016/j.bbrep.2020.100877] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/29/2020] [Accepted: 12/09/2020] [Indexed: 11/12/2022] Open
Abstract
Modern pharmacological studies have demonstrated that Dendrobium nobile Lindl. Alkaloids (DNLA), the main active ingredients of Dendrobium nobile, is valuable as an anti-aging and neuroprotective herbal medicine. The present study was designed to determine whether DNLA confers protective function over neurotoxicant manganese (Mn)-induced cytotoxicity and the mechanism involved. Our results showed that pretreatment of PC12 cells with DNLA alleviated cell toxicity induced by Mn and improved mitochondrial respiratory capacity and oxidative status. Mn treatment increased apoptotic cell death along with a marked increase in the protein expression of Bax and a decrease in the expression of Bcl-2 protein, all of which were noticeably reversed by DNLA. Furthermore, DNLA significantly abolished the decrease in protein levels of both PINK1 and Parkin, and mitigated the increased expression of autophagy marker LC3-II and accumulation of p62 caused by Mn. These results demonstrate that DNLA inhibits Mn induced cytotoxicity, which may be mediated through modulating PINK1/Parkin-mediated autophagic flux and improving mitochondrial function. Dendrobium nobile Lindl. alkaloids (DNLA) significantly alleviate cytotoxicity of PC12 cells induced by manganese (Mn) and improve mitochondrial function. Activation of PINK1/Parkin-mediated autophagic flux is involved in the protective action of DNLA on Mn-induced neurotoxicity. DNLA reduces Mn-induced ROS generation and suppresses Mn-induced apoptosis.
Collapse
Affiliation(s)
- Xiaolong Fu
- Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China.,Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, Guizhou, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, China
| | - Shu Chen
- Cell and Tissue Bank of Guizhou Province, Zunyi, Guizhou, China
| | - Xueting Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China.,Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, Guizhou, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yanhua Shen
- Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China.,Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, Guizhou, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, China
| | - Ru Zeng
- Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China.,Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, Guizhou, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, China
| | - Qin Wu
- Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China.,Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, Guizhou, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yuanfu Lu
- Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China.,Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, Guizhou, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jingshan Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China.,Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, Guizhou, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, China
| | - Shaoyu Zhou
- Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China.,Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, Guizhou, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
21
|
Xu Y, Li R, Li X, Dong N, Wu D, Hou L, Yin K, Zhao C. An Autophagy-Related Gene Signature Associated With Clinical Prognosis and Immune Microenvironment in Gliomas. Front Oncol 2020; 10:571189. [PMID: 33194668 PMCID: PMC7604433 DOI: 10.3389/fonc.2020.571189] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/02/2020] [Indexed: 12/16/2022] Open
Abstract
Glioma is one of the leading causes of death from cancer, and autophagy-related genes (ARGs) play an important role in glioma occurrence, progression, and treatment. In this study, the gene expression profiles and clinical data of glioma patients were obtained from The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA), respectively. ARGs were obtained from the Human Autophagy Database. We analyzed the expression of the ARGs in glioma and found that 73 ARGs were differentially expressed in tumor and normal tissues. Univariate Cox regression analysis was used to identify prognostic differentially expressed ARGs (PDEARGs). Least absolute shrinkage and selection operator (LASSO) and multivariate Cox regression analyses were performed on the PDEARGs to determine the risk genes; and BRIC5, NFE2L2, GABARAP, IKBKE, BID, MAPK3, FKBP1B, MAPK8IP1, PRKCQ, CX3CL1, NPC1, HSP90AB1, DAPK2, SUPT20H, and PTEN were selected to establish a prognostic risk score model for TCGA and CGGA cohorts. This model accurately stratified patients with different survival outcomes, and the autophagy-related signature was also appraised as being an independent prognostic factor. We also constructed a prognostic nomogram using risk score, age, gender, WHO grade, isocitrate dehydrogenase (IDH) mutation status, and 1p/19q co-deletion status; and the calibration plots showed excellent prognostic performance. Finally, Pearson correlation analysis suggested that the ARG signature also played an essential role in the tumor immune microenvironment. In summary, we constructed and verified a novel autophagy-related signature that was tightly associated with the tumor immune microenvironment and could serve as an independent prognostic biomarker in gliomas.
Collapse
Affiliation(s)
- Yang Xu
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Renpeng Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaoxia Li
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Naijun Dong
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Di Wu
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Lin Hou
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Kan Yin
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Chunhua Zhao
- School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
22
|
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
|
23
|
Escamilla-Ramírez A, Castillo-Rodríguez RA, Zavala-Vega S, Jimenez-Farfan D, Anaya-Rubio I, Briseño E, Palencia G, Guevara P, Cruz-Salgado A, Sotelo J, Trejo-Solís C. Autophagy as a Potential Therapy for Malignant Glioma. Pharmaceuticals (Basel) 2020; 13:ph13070156. [PMID: 32707662 PMCID: PMC7407942 DOI: 10.3390/ph13070156] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/01/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
Glioma is the most frequent and aggressive type of brain neoplasm, being anaplastic astrocytoma (AA) and glioblastoma multiforme (GBM), its most malignant forms. The survival rate in patients with these neoplasms is 15 months after diagnosis, despite a diversity of treatments, including surgery, radiation, chemotherapy, and immunotherapy. The resistance of GBM to various therapies is due to a highly mutated genome; these genetic changes induce a de-regulation of several signaling pathways and result in higher cell proliferation rates, angiogenesis, invasion, and a marked resistance to apoptosis; this latter trait is a hallmark of highly invasive tumor cells, such as glioma cells. Due to a defective apoptosis in gliomas, induced autophagic death can be an alternative to remove tumor cells. Paradoxically, however, autophagy in cancer can promote either a cell death or survival. Modulating the autophagic pathway as a death mechanism for cancer cells has prompted the use of both inhibitors and autophagy inducers. The autophagic process, either as a cancer suppressing or inducing mechanism in high-grade gliomas is discussed in this review, along with therapeutic approaches to inhibit or induce autophagy in pre-clinical and clinical studies, aiming to increase the efficiency of conventional treatments to remove glioma neoplastic cells.
Collapse
Affiliation(s)
- Angel Escamilla-Ramírez
- Departamento de Neuroinmunología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de México 14269, Mexico; (A.E.-R.); (I.A.-R.); (G.P.); (P.G.); (A.C.-S.); (J.S.)
| | - Rosa A. Castillo-Rodríguez
- Laboratorio de Oncología Experimental, CONACYT-Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico;
| | - Sergio Zavala-Vega
- Departamento de Patología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de México 14269, Mexico;
| | - Dolores Jimenez-Farfan
- Laboratorio de Inmunología, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Isabel Anaya-Rubio
- Departamento de Neuroinmunología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de México 14269, Mexico; (A.E.-R.); (I.A.-R.); (G.P.); (P.G.); (A.C.-S.); (J.S.)
| | - Eduardo Briseño
- Clínica de Neurooncología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de México 14269, Mexico;
| | - Guadalupe Palencia
- Departamento de Neuroinmunología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de México 14269, Mexico; (A.E.-R.); (I.A.-R.); (G.P.); (P.G.); (A.C.-S.); (J.S.)
| | - Patricia Guevara
- Departamento de Neuroinmunología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de México 14269, Mexico; (A.E.-R.); (I.A.-R.); (G.P.); (P.G.); (A.C.-S.); (J.S.)
| | - Arturo Cruz-Salgado
- Departamento de Neuroinmunología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de México 14269, Mexico; (A.E.-R.); (I.A.-R.); (G.P.); (P.G.); (A.C.-S.); (J.S.)
| | - Julio Sotelo
- Departamento de Neuroinmunología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de México 14269, Mexico; (A.E.-R.); (I.A.-R.); (G.P.); (P.G.); (A.C.-S.); (J.S.)
| | - Cristina Trejo-Solís
- Departamento de Neuroinmunología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de México 14269, Mexico; (A.E.-R.); (I.A.-R.); (G.P.); (P.G.); (A.C.-S.); (J.S.)
- Correspondence: ; Tel.: +52-555-060-4040
| |
Collapse
|
24
|
Wang QW, Liu HJ, Zhao Z, Zhang Y, Wang Z, Jiang T, Bao ZS. Prognostic Correlation of Autophagy-Related Gene Expression-Based Risk Signature in Patients with Glioblastoma. Onco Targets Ther 2020; 13:95-107. [PMID: 32021258 PMCID: PMC6954841 DOI: 10.2147/ott.s238332] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 12/17/2019] [Indexed: 12/21/2022] Open
Abstract
Purpose Autophagy plays a vital role in cancer initiation, malignant progression, and resistance to treatment; however, autophagy-related gene sets have rarely been analyzed in glioblastoma. The purpose of this study was to evaluate the prognostic significance of autophagy-related genes in patients with glioblastoma. Patients and methods Here, we collected whole transcriptome expression data from the Chinese Glioma Genome Atlas (CGGA) and The Cancer Genome Atlas (TCGA) datasets to explore the relationship between autophagy-related gene expression and glioblastoma prognosis. R language was the primary analysis and drawing tool. Results We screened 531 autophagy-related genes and identified 14 associated with overall survival in data from 986 patients with glioblastoma. Patients could be clustered into two groups (high and low risk) using expression data from the 14 associated genes, based on significant differences in clinicopathology and prognosis. Next, we constructed a signature based on the 14 genes and found that most patients designated high risk using our gene signature were IDH wild-type, MGMT promoter non-methylated, and likely to have more malignant tumor subtypes (including classical and mesenchymal subtypes). Survival analysis indicated that patients in the high-risk group had dramatically shorter overall survival compared with their low-risk counterparts. Cox regression analysis further confirmed the independent prognostic value of our 14 gene signature. Moreover, functional and ESTIMATE analyses revealed enrichment of immune and inflammatory responses in the high-risk group. Conclusion In this study, we identified a novel autophagy-related signature for the prediction of prognosis in patients with glioblastoma.
Collapse
Affiliation(s)
- Qiang-Wei Wang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, People's Republic of China
| | - Han-Jie Liu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, People's Republic of China
| | - Zheng Zhao
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, People's Republic of China
| | - Ying Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, People's Republic of China
| | - Zheng Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, People's Republic of China
| | - Tao Jiang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, People's Republic of China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, People's Republic of China
| | - Zhao-Shi Bao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, People's Republic of China.,Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, People's Republic of China
| |
Collapse
|
25
|
Petővári G, Dankó T, Krencz I, Hujber Z, Rajnai H, Vetlényi E, Raffay R, Pápay J, Jeney A, Sebestyén A. Inhibition of Metabolic Shift can Decrease Therapy Resistance in Human High-Grade Glioma Cells. Pathol Oncol Res 2020; 26:23-33. [PMID: 31187466 PMCID: PMC7109188 DOI: 10.1007/s12253-019-00677-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 05/28/2019] [Indexed: 12/19/2022]
Abstract
The high-grade brain malignancy, glioblastoma multiforme (GBM), is one of the most aggressive tumours in central nervous system. The developing resistance against recent therapies and the recurrence rate of GBMs are extremely high. In spite several new ongoing trials, GBM therapies could not significantly increase the survival rate of the patients as significantly. The presence of inter- and intra-tumoral heterogeneity of GBMs arise the problem to find both the pre-existing potential resistant clones and the cellular processes which promote the adaptation mechanisms such as multidrug resistance, stem cell-ness or metabolic alterations, etc. In our work, the in situ metabolic heterogeneity of high-grade human glioblastoma cases were analysed by immunohistochemistry using tissue-microarray. The potential importance of the detected metabolic heterogeneity was tested in three glioma cell lines (grade III-IV) using protein expression analyses (Western blot and WES Simple) and therapeutic drug (temozolomide), metabolic inhibitor treatments (including glutaminase inhibitor) to compare the effects of rapamycin (RAPA) and glutaminase inhibitor combinations in vitro (Alamar Blue and SRB tests). The importance of individual differences and metabolic alterations were observed in mono-therapeutic failures, especially the enhanced Rictor expressions after different mono-treatments in correlation to lower sensitivity (temozolomide, doxycycline, etomoxir, BPTES). RAPA combinations with other metabolic inhibitors were the best strategies except for RAPA+glutaminase inhibitor. These observations underline the importance of multi-targeting metabolic pathways. Finally, our data suggest that the detected metabolic heterogeneity (the high mTORC2 complex activity, enhanced expression of Rictor, p-Akt, p-S6, CPT1A, and LDHA enzymes in glioma cases) and the microenvironmental or treatment induced metabolic shift can be potential targets in combination therapy. Therefore, it should be considered to map tissue heterogeneity and alterations with several cellular metabolism markers in biopsy materials after applying recently available or new treatments.
Collapse
Affiliation(s)
- Gábor Petővári
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Titanilla Dankó
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Ildikó Krencz
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Zoltán Hujber
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Hajnalka Rajnai
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Enikő Vetlényi
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Regina Raffay
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Judit Pápay
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - András Jeney
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Anna Sebestyén
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary.
| |
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
|
Palumbo P, Lombardi F, Augello FR, Giusti I, Luzzi S, Dolo V, Cifone MG, Cinque B. NOS2 inhibitor 1400W Induces Autophagic Flux and Influences Extracellular Vesicle Profile in Human Glioblastoma U87MG Cell Line. Int J Mol Sci 2019; 20:ijms20123010. [PMID: 31226744 PMCID: PMC6627770 DOI: 10.3390/ijms20123010] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 06/13/2019] [Accepted: 06/17/2019] [Indexed: 12/29/2022] Open
Abstract
The relevance of nitric oxide synthase 2 (NOS2) as a prognostic factor in Glioblastoma Multiforme (GBM) malignancy is emerging. We analyzed the effect of NOS2 inhibitor 1400W on the autophagic flux and extracellular vesicle (EV) secretion in U87MG glioma cells. The effects of glioma stem cells (GSC)-derived EVs on adherent U87MG were evaluated. Cell proliferation and migration were examined while using Cell Counting Kit-8 assay (CCK-8) and scratch wound healing assay. Cell cycle profile and apoptosis were analyzed by flow cytometry. Autophagy-associated acidic vesicular organelles were detected and quantified by acridine orange staining. The number and size of EVs were assessed by nanoparticle tracking analysis. EV ultrastructure was verified by transmission electron microscopy (TEM). WB was used to analyze protein expression and acid sphingomyelinase was determined through ceramide levels. 1400W induced autophagy and EV secretion in both adherent U87MG and GSCs. EVs secreted by 1400W-treated GSC, but not those from untreated cells, were able to inhibit adherent U87MG cell growth and migration while also inducing a relevant level of autophagy. The hypothesis of NOS2 expression as GBM profile marker or interesting therapeutic target is supported by our findings. Autophagy and EV release following treatment with the NOS2 inhibitor could represent useful elements to better understand the complex biomolecular frame of GBM.
Collapse
Affiliation(s)
- Paola Palumbo
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, Coppito, 67100 L'Aquila, Italy.
| | - Francesca Lombardi
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, Coppito, 67100 L'Aquila, Italy.
| | - Francesca Rosaria Augello
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, Coppito, 67100 L'Aquila, Italy.
| | - Ilaria Giusti
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, Coppito, 67100 L'Aquila, Italy.
| | - Sabino Luzzi
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Polo Didattico "Cesare Brusotti", Viale Brambilla, 74 - 27100 Pavia, Italy.
| | - Vincenza Dolo
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, Coppito, 67100 L'Aquila, Italy.
| | - Maria Grazia Cifone
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, Coppito, 67100 L'Aquila, Italy.
| | - Benedetta Cinque
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, Coppito, 67100 L'Aquila, Italy.
| |
Collapse
|
28
|
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
|
29
|
Park HS, Han JH, Jung SH, Lee DH, Heo KS, Myung CS. Anti-apoptotic effects of autophagy via ROS regulation in microtubule-targeted and PDGF-stimulated vascular smooth muscle cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2018; 22:349-360. [PMID: 29719457 PMCID: PMC5928348 DOI: 10.4196/kjpp.2018.22.3.349] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 03/10/2018] [Accepted: 03/11/2018] [Indexed: 01/09/2023]
Abstract
Autophagy has been studied as a therapeutic strategy for cardiovascular diseases. However, insufficient studies have been reported concerning the influence of vascular smooth muscle cells (VSMCs) through autophagy regulation. The aim of the present study was to determine the effects of VSMCs on the regulation of autophagy under in vitro conditions similar to vascular status of the equipped microtubule target agent-eluting stent and increased release of platelet-derived growth factor-BB (PDGF-BB). Cell viability and proliferation were measured using MTT and cell counting assays. Immunofluorescence using an anti-α-tubulin antibody was performed to determine microtubule dynamic formation. Cell apoptosis was measured by cleavage of caspase-3 using western blot analysis, and by nuclear fragmentation using a fluorescence assay. Autophagy activity was assessed by microtubule-associated protein light chain 3-II (LC-II) using western blot analysis. Levels of intracellular reactive oxygen species (ROS) were measured using H2DCFDA. The proliferation and viability of VSMCs were inhibited by microtubule regulation. Additionally, microtubule-regulated and PDGF-BB-stimulated VSMCs increased the cleavage of caspase-3 more than only the microtubule-regulated condition, similar to that of LC3-II, implying autophagy. Inhibitory autophagy of microtubule-regulated and PDGF-BB-stimulated VSMCs resulted in low viability. However, enhancement of autophagy maintained survival through the reduction of ROS. These results suggest that the apoptosis of conditioned VSMCs is decreased by the blocking generation of ROS via the promotion of autophagy, and proliferation is also inhibited. Thus, promoting autophagy as a therapeutic target for vascular restenosis and atherosclerosis may be a good strategy.
Collapse
Affiliation(s)
- Hyun-Soo Park
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 34134, Korea
| | - Joo-Hui Han
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 34134, Korea
| | - Sang-Hyuk Jung
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 34134, Korea
| | - Do-Hyung Lee
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 34134, Korea
| | - Kyung-Sun Heo
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 34134, Korea
| | - Chang-Seon Myung
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 34134, Korea.,Institute of Drug Research & Development, Chungnam National University, Daejeon 34134, Korea
| |
Collapse
|