51
|
Stanković T, Ranđelović T, Dragoj M, Stojković Burić S, Fernández L, Ochoa I, Pérez-García VM, Pešić M. In vitro biomimetic models for glioblastoma-a promising tool for drug response studies. Drug Resist Updat 2021; 55:100753. [PMID: 33667959 DOI: 10.1016/j.drup.2021.100753] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023]
Abstract
The poor response of glioblastoma to current treatment protocols is a consequence of its intrinsic drug resistance. Resistance to chemotherapy is primarily associated with considerable cellular heterogeneity, and plasticity of glioblastoma cells, alterations in gene expression, presence of specific tumor microenvironment conditions and blood-brain barrier. In an attempt to successfully overcome chemoresistance and better understand the biological behavior of glioblastoma, numerous tri-dimensional (3D) biomimetic models were developed in the past decade. These novel advanced models are able to better recapitulate the spatial organization of glioblastoma in a real time, therefore providing more realistic and reliable evidence to the response of glioblastoma to therapy. Moreover, these models enable the fine-tuning of different tumor microenvironment conditions and facilitate studies on the effects of the tumor microenvironment on glioblastoma chemoresistance. This review outlines current knowledge on the essence of glioblastoma chemoresistance and describes the progress achieved by 3D biomimetic models. Moreover, comprehensive literature assessment regarding the influence of 3D culturing and microenvironment mimicking on glioblastoma gene expression and biological behavior is also provided. The contribution of the blood-brain barrier as well as the blood-tumor barrier to glioblastoma chemoresistance is also reviewed from the perspective of 3D biomimetic models. Finally, the role of mathematical models in predicting 3D glioblastoma behavior and drug response is elaborated. In the future, technological innovations along with mathematical simulations should create reliable 3D biomimetic systems for glioblastoma research that should facilitate the identification and possibly application in preclinical drug testing and precision medicine.
Collapse
Affiliation(s)
- Tijana Stanković
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060, Belgrade, Serbia
| | - Teodora Ranđelović
- Tissue Microenvironment Lab (TME), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Aragon 50018, Spain; Institute for Health Research Aragon (IIS Aragón), Instituto de Salud Carlos III, Zaragoza, Spain
| | - Miodrag Dragoj
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060, Belgrade, Serbia
| | - Sonja Stojković Burić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060, Belgrade, Serbia
| | - Luis Fernández
- Tissue Microenvironment Lab (TME), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Aragon 50018, Spain; Centro Investigación Biomédica en Red. Bioingenieria, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, Aragon 50018, Spain; Institute for Health Research Aragon (IIS Aragón), Instituto de Salud Carlos III, Zaragoza, Spain
| | - Ignacio Ochoa
- Tissue Microenvironment Lab (TME), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Aragon 50018, Spain; Centro Investigación Biomédica en Red. Bioingenieria, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, Aragon 50018, Spain; Institute for Health Research Aragon (IIS Aragón), Instituto de Salud Carlos III, Zaragoza, Spain
| | - Victor M Pérez-García
- Departamento de Matemáticas, E.T.S.I. Industriales and Instituto de Matemática Aplicada a la Ciencia y la Ingeniería (IMACI), Universidad de Castilla-La Mancha, Ciudad Real, 13071, Spain
| | - Milica Pešić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060, Belgrade, Serbia.
| |
Collapse
|
52
|
Li L, Wang F, Zhang J, Wang K, De X, Li L, Zhang Y. Typical phthalic acid esters induce apoptosis by regulating the PI3K/Akt/Bcl-2 signaling pathway in rat insulinoma cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 208:111461. [PMID: 33091774 DOI: 10.1016/j.ecoenv.2020.111461] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 07/29/2020] [Accepted: 10/01/2020] [Indexed: 06/11/2023]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) and dibutyl phthalate (DBP) are representative phthalic acid esters (PAEs), a class of environmental endocrine disruptors used as plasticizers. PAEs exposure is associated with glucose metabolism, insulin resistance, and glucose tolerance; however, the mechanism and various PAE effects on human glucose metabolism remain largely unknown. In this study, we investigated the effects of DEHP, DBP, and their mixture on rat insulinoma (INS-1) cell apoptosis and the mechanism involved in vitro. The INS-1 cells were cultured in RPMI-1640 + 10% fetal bovine serum for 24 h and pretreated with dimethyl sulfoxide (vehicle, <0.1%), DEHP (30 μM), DBP (30 μM), and their mixture (30 μM DEHP + 30 μM DBP). The methyl-thiazolyl tetrazolium bromide test was used to measure cell viability. Hoechst 33342/propidium iodide (PI) staining and Annexin V-FITC/PI staining, 2',7'-dichlorofluorescein diacetate assay, and glucose-induced insulin secretion assay were used to detect cell apoptosis rates, intracellular reactive oxygen species (ROS), and insulin secretion in INS-1, respectively. The mRNA expression levels of Bcl-2, Bax, Caspase 9, Caspase 8, Caspase 3, phosphoinositide 3-kinase (PI3K), and Akt were detected using real-time quantitative reverse transcription PCR; their protein expression levels were detected using western blotting. To the best of our knowledge, this study was the first to show that the combined effect of the two PAEs promotes a ROS-mediated PI3K/Akt/Bcl-2 pathway-induced pancreatic β cell apoptosis that is significantly higher than the effects of each PAE. Thus, safety standards and studies do not consider this effect as a significant oversight when blending PAEs. We assert that this must be addressed and corrected for establishing more impactful and safer standards.
Collapse
Affiliation(s)
- Liping Li
- School of Public Health and Management, Ningxia Medical University, Yinchuan 750004, Ningxia, China; Key Laboratory of Fertility Preservation and Maintenance of the Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia, China.
| | - Faxuan Wang
- School of Public Health and Management, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Jianjun Zhang
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, Indiana, United States.
| | - Kai Wang
- School of Public Health and Management, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Xiaoming De
- School of Public Health and Management, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Ling Li
- School of Public Health and Management, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Yuhong Zhang
- School of Public Health and Management, Ningxia Medical University, Yinchuan 750004, Ningxia, China; Key Laboratory of Fertility Preservation and Maintenance of the Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia, China.
| |
Collapse
|
53
|
Ma X, Yu M, Hao C, Yang W. Shikonin induces tumor apoptosis in glioma cells via endoplasmic reticulum stress, and Bax/Bak mediated mitochondrial outer membrane permeability. JOURNAL OF ETHNOPHARMACOLOGY 2020; 263:113059. [PMID: 32663591 DOI: 10.1016/j.jep.2020.113059] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 04/09/2020] [Accepted: 05/31/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shikonin, one of the main active ingredients of Chinese herbal medicine Lithospermum erythrorhizon, has been widely used to treat various disease including virus infection and inflammation in clinical. Its anti-tumor activity has been recorded in "Chinese herbal medicine". Recently, some studies about its anti-glioma effects have been reported. However, little is known about the molecular pharmacological activity of Shikonin in glioma. AIM This study aimed to systematically uncover and validate the pharmacological mechanism of Shikonin against glioma. MATERIAL AND METHODS Network pharmacology approach, survival analysis, and Pearson co-expression analysis were performed to uncover and test the pharmacological mechanisms of Shikonin in glioma. Apoptosis assay, Caspase-3 activity assay and immunoblot analysis were practiced to validate the mechanisms. RESULTS Network pharmacology results suggested, anti-glioma effect of Shikonin by interfering endoplasmic reticulum (ER) stress-mediated tumor apoptosis targeting Caspase-3, and Bax/Bak-induced mitochondrial outer membrane permeabilization (MOMP) triggering cancer cell apoptosis. Survival analysis suggested the association of CASP3 with glioma (P < 0.05). Pearson correlation analysis indicated possible interaction of CASP3 with PERK through positive feedback regulation. Shikonin or in combination with 14G2a induced cell apoptosis in oligodendroglioma Hs683 cells in a dose-dependent manner with at a maximum apoptosis rate of 33%-37.5%, and 73%-77% respectively. Immunoblot analysis showed that Shikonin increased Caspase-3 activity to about 4.29 times, and increased 9 times when it combined with 14G2a. Shikonin increased also the expression levels of the proteins PERK and CHOP by about 4.4 and 5.6 folds, respectively, when it combined with 14G2a. CONCLUSIONS This study highlights the pharmacological mechanisms of Shikonin in the induction of tumor apoptosis in glioma cells.
Collapse
Affiliation(s)
- Xiaoqin Ma
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Meixiang Yu
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Chenxia Hao
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Wanhua Yang
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
54
|
Xia Q, Xu M, Zhang P, Liu L, Meng X, Dong L. Therapeutic Potential of Autophagy in Glioblastoma Treatment With Phosphoinositide 3-Kinase/Protein Kinase B/Mammalian Target of Rapamycin Signaling Pathway Inhibitors. Front Oncol 2020; 10:572904. [PMID: 33123479 PMCID: PMC7567033 DOI: 10.3389/fonc.2020.572904] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/20/2020] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GB) is the most malignant and aggressive form of brain tumor, characterized by frequent hyperactivation of the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway. PI3K/AKT/mTOR inhibitors have a promising clinical efficacy theoretically. However, strong drug resistance is developed in GB against the PI3K/AKT/mTOR inhibitors due to the cytoprotective effect and the adaptive response of autophagy during the treatment of GB. Activation of autophagy by the PI3K/AKT/mTOR inhibitors not only enhances treatment sensitivity but also leads to cell survival when drug resistance develops in cancer cells. In this review, we analyze how to increase the antitumor effect of the PI3K/AKT/mTOR inhibitors in GB treatment, which is achieved by various mechanisms, among which targeting autophagy is an important mechanism. We review the dual role of autophagy in both GB therapy and resistance against inhibitors of the PI3K/AKT/mTOR signaling pathway, and further discuss the possibility of using combinations of autophagy and PI3K/AKT/mTOR inhibitors to improve the treatment efficacy for GB. Finally, we provide new perspectives for targeting autophagy in GB therapy.
Collapse
Affiliation(s)
- Qin Xia
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Mengchuan Xu
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Pei Zhang
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Liqun Liu
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Xinyi Meng
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Lei Dong
- School of Life Science, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
55
|
Hwang M, Park HH, Han MH, Choi H, Lee KY, Lee YJ, Kim JM, Cheong JH, Ryu JI, Ko Y, Koh SH. Chemoradiotherapy Alters Protein Expression in Glioblastoma Multiforme. J Clin Neurol 2020; 16:725-728. [PMID: 33029989 PMCID: PMC7541983 DOI: 10.3988/jcn.2020.16.4.725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 08/09/2020] [Accepted: 08/12/2020] [Indexed: 11/22/2022] Open
Affiliation(s)
- Mina Hwang
- Department of Neurology, Hanyang University Guri Hospital, Guri, Korea
| | - Hyun Hee Park
- Department of Neurology, Hanyang University Guri Hospital, Guri, Korea
| | - Myung Hoon Han
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri, Korea.
| | - Hojin Choi
- Department of Neurology, Hanyang University Guri Hospital, Guri, Korea
| | - Kyu Yong Lee
- Department of Neurology, Hanyang University Guri Hospital, Guri, Korea
| | - Young Joo Lee
- Department of Neurology, Hanyang University Guri Hospital, Guri, Korea
| | - Jae Min Kim
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri, Korea
| | - Jin Hwan Cheong
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri, Korea
| | - Je Il Ryu
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri, Korea
| | - Yong Ko
- Department of Neurosurgery, Hanyang University Medical Center, Seoul, Korea
| | - Seong Ho Koh
- Department of Neurology, Hanyang University Guri Hospital, Guri, Korea
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul, Korea.
| |
Collapse
|
56
|
Wehbe N, Slika H, Mesmar J, Nasser SA, Pintus G, Baydoun S, Badran A, Kobeissy F, Eid AH, Baydoun E. The Role of Epac in Cancer Progression. Int J Mol Sci 2020; 21:ijms21186489. [PMID: 32899451 PMCID: PMC7555121 DOI: 10.3390/ijms21186489] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer continues to be a prime contributor to global mortality. Despite tremendous research efforts and major advances in cancer therapy, much remains to be learned about the underlying molecular mechanisms of this debilitating disease. A better understanding of the key signaling events driving the malignant phenotype of cancer cells may help identify new pharmaco-targets. Cyclic adenosine 3',5'-monophosphate (cAMP) modulates a plethora of biological processes, including those that are characteristic of malignant cells. Over the years, most cAMP-mediated actions were attributed to the activity of its effector protein kinase A (PKA). However, studies have revealed an important role for the exchange protein activated by cAMP (Epac) as another effector mediating the actions of cAMP. In cancer, Epac appears to have a dual role in regulating cellular processes that are essential for carcinogenesis. In addition, the development of Epac modulators offered new routes to further explore the role of this cAMP effector and its downstream pathways in cancer. In this review, the potentials of Epac as an attractive target in the fight against cancer are depicted. Additionally, the role of Epac in cancer progression, namely its effect on cancer cell proliferation, migration/metastasis, and apoptosis, with the possible interaction of reactive oxygen species (ROS) in these phenomena, is discussed with emphasis on the underlying mechanisms and pathways.
Collapse
Affiliation(s)
- Nadine Wehbe
- Department of Biology, American University of Beirut, P.O. Box 11-0236 Beirut, Lebanon; (N.W.); (J.M.)
| | - Hasan Slika
- Department of Pharmacology and Therapeutics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236 Beirut, Lebanon;
| | - Joelle Mesmar
- Department of Biology, American University of Beirut, P.O. Box 11-0236 Beirut, Lebanon; (N.W.); (J.M.)
| | - Suzanne A. Nasser
- Department of Pharmacology, Beirut Arab University, P.O. Box 11-5020 Beirut, Lebanon;
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sharjah, P.O. Box 27272 Sharjah, UAE;
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100 Sassari, Italy
| | - Serine Baydoun
- Department of Radiology, American University of Beirut, P.O. Box 11-0236 Beirut, Lebanon;
| | - Adnan Badran
- Department of Basic Sciences, University of Petra, P.O. Box 961343, Amman 11196, Jordan;
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon;
| | - Ali H. Eid
- Department of Pharmacology and Therapeutics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236 Beirut, Lebanon;
- Department of Pharmacology and Therapeutics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon
- Correspondence: (A.H.E.); (E.B.); Tel.: +961-1-350-000 (ext. 4891) (A.H.E. & E.B.)
| | - Elias Baydoun
- Department of Biology, American University of Beirut, P.O. Box 11-0236 Beirut, Lebanon; (N.W.); (J.M.)
- Correspondence: (A.H.E.); (E.B.); Tel.: +961-1-350-000 (ext. 4891) (A.H.E. & E.B.)
| |
Collapse
|
57
|
Liu W, Zhou J, Niu F, Pu F, Wang Z, Huang M, Zhao X, Yang L, Tao P, Xia P, Feng J. Mycobacterium tuberculosis infection increases the number of osteoclasts and inhibits osteoclast apoptosis by regulating TNF-α-mediated osteoclast autophagy. Exp Ther Med 2020; 20:1889-1898. [PMID: 32782497 PMCID: PMC7401307 DOI: 10.3892/etm.2020.8903] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 12/11/2019] [Indexed: 02/07/2023] Open
Abstract
Osteoarticular tuberculosis, a chronic inflammatory disease characterized by Mycobacterium tuberculosis (M.tb) infection, has become a serious problem in China. The present study was conducted to determine the mechanism of action of tumor necrosis factor (TNF)-α in the pathogenesis of osteoarticular tuberculosis. The number of osteoclasts in osteoarticular tuberculosis tissue samples was detected by tartrate-resistant acid phosphatase staining. Autophagy and apoptosis of osteoclasts were detected by western blotting, reverse transcription-quantitative PCR, transmission electron microscopy and TUNEL staining. The results showed that autophagy and the number of osteoclasts increased in the lesions of patients with osteoarticular tuberculosis compared with osteoarthritis samples. Moreover, activation of osteoclast autophagy inhibited the apoptosis of osteoclasts infected with M.tb, and increased the expression level of TNF-α. The results showed that TNF-α enhanced the autophagic activity of M.tb-infected osteoclasts and inhibited cell apoptosis. These findings indicated that M.tb infection induced osteoclast production and inhibited osteoclast apoptosis by regulating TNF-α-mediated osteoclast autophagy, revealing a new mechanism for TNF-α in the pathogenesis of osteoarticular tuberculosis.
Collapse
Affiliation(s)
- Wei Liu
- Department of Orthopaedics, First Hospital of Wuhan, Wuhan, Hubei 430022, P.R. China
| | - Juan Zhou
- Department of Orthopaedics, First Hospital of Wuhan, Wuhan, Hubei 430022, P.R. China
| | - Fei Niu
- Department of Orthopaedics, First Hospital of Wuhan, Wuhan, Hubei 430022, P.R. China
| | - Feifei Pu
- Department of Orthopaedics, First Hospital of Wuhan, Wuhan, Hubei 430022, P.R. China
| | - Zhiwei Wang
- Department of Orthopaedics, First Hospital of Wuhan, Wuhan, Hubei 430022, P.R. China
| | - Mi Huang
- Department of Orthopaedics, First Hospital of Wuhan, Wuhan, Hubei 430022, P.R. China
| | - Xiaolong Zhao
- Department of Orthopaedics, First Hospital of Wuhan, Wuhan, Hubei 430022, P.R. China
| | - Lin Yang
- Department of Orthopaedics, First Hospital of Wuhan, Wuhan, Hubei 430022, P.R. China
| | - Pengfei Tao
- Department of Orthopaedics, First Hospital of Wuhan, Wuhan, Hubei 430022, P.R. China
| | - Ping Xia
- Department of Orthopaedics, First Hospital of Wuhan, Wuhan, Hubei 430022, P.R. China
| | - Jing Feng
- Department of Orthopaedics, First Hospital of Wuhan, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
58
|
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: 1.3] [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
|
59
|
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: 52] [Impact Index Per Article: 13.0] [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
|
60
|
Wang P, Wang X, Tang Q, Chen H, Zhang Q, Jiang H, Wang Z. Functionalized graphene oxide against U251 glioma cells and its molecular mechanism. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 116:111187. [PMID: 32806260 DOI: 10.1016/j.msec.2020.111187] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/22/2020] [Accepted: 06/10/2020] [Indexed: 12/20/2022]
Abstract
Graphene and its derivatives with exceptional properties are being exploited for drug delivery and even combined therapies for enhanced antitumor activity and reduced side effects. However, the unfavorable surface chemistry of pristine graphene and reduced graphene oxide made them take covalent and non-covalent functionalization strategies to improve their biocompatibility. Although graphene oxide (GO) is soluble in water owing to its oxygen-containing groups such as carboxylic acid and hydroxyl groups, it is highly accepted when to be modified to improve its colloidal stability in physiological buffers in the presence of salts. In this work, we functionalized GO with Pluronic F127 molecules via non-covalent interaction and found that GO and PF127/GO nanohybrid with a concentration lower than 5 μg/ml have no obvious toxic effect on human astrocytes (AS) and human glioma (U251) cells. Anti-tumor drug doxorubicin (DOX) being loaded onto the PF127/GO nanocarriers by π-π stacking exhibited a high loading capacity of 0.83 mg/mg and loading efficiency of 83%. Our study confirmed that the PF127/GO/DOX (PGD) induced a higher apoptosis rate (12.27 ± 0.06%) of U251 cells than that of free DOX (8.20 ± 0.06%) (P < 0.05). Western blotting results indicated that PGD affected the MAPK signaling pathway and induced the intrinsic pathway of apoptosis for the activation of Caspase-3 in U251 cells, which may provide more evidence for the signal pathway of tumor-targeting therapy.
Collapse
Affiliation(s)
- Pingyue Wang
- Neurology Department, The First Hospital of Jilin University, Changchun 130021, China
| | - Xin Wang
- Key Laboratory of Automobile Materials of MOE, College of Materials Science and Engineering, Jilin University, Changchun 130012, China.
| | - Qi Tang
- Neurology Department, The First Hospital of Jilin University, Changchun 130021, China
| | - Hao Chen
- Key Laboratory of Automobile Materials of MOE, College of Materials Science and Engineering, Jilin University, Changchun 130012, China
| | - Qin Zhang
- Neurology Department, The First Hospital of Jilin University, Changchun 130021, China
| | - Hongyu Jiang
- Department of Health Examination Centre, The First Hospital of Jilin University, Changchun 130021, China
| | - Zan Wang
- Neurology Department, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
61
|
Palumbo P, Lombardi F, Augello FR, Giusti I, Dolo V, Leocata P, Cifone MG, Cinque B. Biological effects of selective COX-2 inhibitor NS398 on human glioblastoma cell lines. Cancer Cell Int 2020; 20:167. [PMID: 32435158 PMCID: PMC7222447 DOI: 10.1186/s12935-020-01250-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/07/2020] [Indexed: 12/14/2022] Open
Abstract
Background Cyclooxygenase-2 (COX-2), an inflammation-associated enzyme, has been implicated in tumorigenesis and progression of glioblastoma (GBM). The poor survival of GBM was mainly associated with the presence of glioma stem cells (GSC) and the markedly inflammatory microenvironment. To further explore the involvement of COX-2 in glioma biology, the effects of NS398, a selective COX-2 inhibitor, were evaluated on GSC derived from COX-2 expressing GBM cell lines, i.e., U87MG and T98G, in terms of neurospheres' growth, autophagy, and extracellular vesicle (EV) release. Methods Neurospheres' growth and morphology were evaluated by optical and scanning electron microscopy. Autophagy was measured by staining acidic vesicular organelles. Extracellular vesicles (EV), released from neurospheres, were analyzed by transmission electron microscopy. The autophagic proteins Beclin-1 and LC3B, as well as the EV markers CD63 and CD81, were analyzed by western blotting. The scratch assay test was used to evaluate the NS398 influence on GBM cell migration. Results Both cell lines were strongly influenced by NS398 exposure, as showed by morphological changes, reduced growth rate, and appearance of autophagy. Furthermore, the inhibitor led to a functional change of EV released by neurospheres. Indeed, EV secreted by NS398-treated GSC, but not those from control cells, were able to significantly inhibit adherent U87MG and T98G cell migration and induced autophagy in recipient cells, thus leading to effects quite similar to those directly caused by NS398 in the same cells. Conclusion Despite the intrinsic diversity and individual genetic features of U87MG and T98G, comparable effects were exerted by the COX-2 inhibitor NS398 on both GBM cell lines. Overall, our findings support the crucial role of the inflammatory-associated COX-2/PGE2 system in glioma and glioma stem cell biology.
Collapse
Affiliation(s)
- Paola Palumbo
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Francesca Lombardi
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | | | - Ilaria Giusti
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Vincenza Dolo
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Pietro Leocata
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Maria Grazia Cifone
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Benedetta Cinque
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| |
Collapse
|
62
|
Wang Z, Gao L, Guo X, Feng C, Lian W, Deng K, Xing B. Development and validation of a nomogram with an autophagy-related gene signature for predicting survival in patients with glioblastoma. Aging (Albany NY) 2019; 11:12246-12269. [PMID: 31844032 PMCID: PMC6949068 DOI: 10.18632/aging.102566] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 11/20/2019] [Indexed: 01/08/2023]
Abstract
Glioblastoma (GBM) is the most common brain tumor with significant morbidity and mortality. Autophagy plays a vital role in GBM development and progression. We aimed to establish an autophagy-related multigene expression signature for individualized prognosis prediction in patients with GBM. Differentially expressed autophagy-related genes (DE-ATGs) in GBM and normal samples were screened using TCGA. Univariate and multivariate Cox regression analyses were performed on DE-ATGs to identify the optimal prognosis-related genes. Consequently, NRG1 (HR=1.142, P=0.008), ITGA3 (HR=1.149, P=0.043), and MAP1LC3A (HR=1.308, P=0.014) were selected to establish the prognostic risk score model and validated in the CGGA validation cohort. GSEA revealed that these genes were mainly enriched in cancer- and autophagy-related KEGG pathways. Kaplan-Meier survival analysis demonstrated that patients with high risk scores had significantly poorer overall survival (OS, log-rank P= 6.955×10-5). The autophagy signature was identified as an independent prognostic factor. Finally, a prognostic nomogram including the autophagy signature, age, pharmacotherapy, radiotherapy, and IDH mutation status was constructed, and TCGA/CGGA-based calibration plots indicated its excellent predictive performance. The autophagy-related three-gene risk score model could be a prognostic biomarker and suggest therapeutic targets for GBM. The prognostic nomogram could assist individualized survival prediction and improve treatment strategies.
Collapse
Affiliation(s)
- Zihao Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng, Beijing 100730, P.R. China.,China Pituitary Disease Registry Center, Chinese Pituitary Adenoma Cooperative Group, Dongcheng, Beijing 100730, P.R. China
| | - Lu Gao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng, Beijing 100730, P.R. China.,China Pituitary Disease Registry Center, Chinese Pituitary Adenoma Cooperative Group, Dongcheng, Beijing 100730, P.R. China
| | - Xiaopeng Guo
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng, Beijing 100730, P.R. China.,China Pituitary Disease Registry Center, Chinese Pituitary Adenoma Cooperative Group, Dongcheng, Beijing 100730, P.R. China
| | - Chenzhe Feng
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng, Beijing 100730, P.R. China.,China Pituitary Disease Registry Center, Chinese Pituitary Adenoma Cooperative Group, Dongcheng, Beijing 100730, P.R. China
| | - Wei Lian
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng, Beijing 100730, P.R. China.,China Pituitary Disease Registry Center, Chinese Pituitary Adenoma Cooperative Group, Dongcheng, Beijing 100730, P.R. China
| | - Kan Deng
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng, Beijing 100730, P.R. China.,China Pituitary Disease Registry Center, Chinese Pituitary Adenoma Cooperative Group, Dongcheng, Beijing 100730, P.R. China
| | - Bing Xing
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng, Beijing 100730, P.R. China.,China Pituitary Disease Registry Center, Chinese Pituitary Adenoma Cooperative Group, Dongcheng, Beijing 100730, P.R. China
| |
Collapse
|
63
|
Wang M, Wu Q, Fang M, Huang W, Zhu H. miR-152-3p Sensitizes Glioblastoma Cells Towards Cisplatin Via Regulation Of SOS1. Onco Targets Ther 2019; 12:9513-9525. [PMID: 31807027 PMCID: PMC6857816 DOI: 10.2147/ott.s210732] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 09/23/2019] [Indexed: 12/24/2022] Open
Abstract
Background Accumulating evidences suggest that microRNAs (miRNAs) play key roles in mediating glioblastoma progression. Decreased expression of miR-152-3p was reported in several cancer types including glioblastoma. Methods The sensitivity of glioblastoma cells to cisplatin was assessed by the cell counting kit-8 assay and flow cytometry analysis. The expression of miR-152-3p was determined by RT-qPCR method. Bioinformatic analysis, dual luciferase reporter assay and Western blot were used to explore the target gene of miR-152-3p. The association between miR-152-3p and SOS1 was confirmed in glioblastoma tissues by Pearson correlation analysis. Results In the current study, we discovered that overexpression of miR-152-3p increased cisplatin sensitivity while inhibition of miR-152-3p decreased cisplatin sensitivity in glioblastoma cells (T98G and U87). In addition, miR-152-3p augmented cell apoptosis induced by cisplatin treatment. It was further predicted and validated that SOS1, a protein involved in regulating chemotherapy sensitivity, was a direct target gene of miR-152-3p. SOS1 was proven to suppress the cytotoxic effect of cisplatin in glioblastoma. Transfection of recombinant SOS1 could effectively reverse the increased cisplatin sensitivity induced by miR-152-3p overexpression in T98G. Furthermore, overexpression of SOS1 reduced the percentage of apoptotic cells increased by miR-152-3p mimic in the presence of cisplatin in T98G. More importantly, a significant negative correlation between miR-152-3p levels and SOS1 levels was observed in glioblastoma tissues collected from 40 patients. Conclusion Our study identified miR-152-3p as a chemotherapy sensitizer in glioblastoma.
Collapse
Affiliation(s)
- Meihua Wang
- Department of Pathology, Changzhou Tumor Hospital, Affiliated to Soochow University, Changzhou, People's Republic of China
| | - Qi Wu
- Department of Histology and Embryology, Heze Medical College, Heze, People's Republic of China
| | - Mingming Fang
- Department of Radiotherapy, Changzhou Tumor Hospital, Affiliated to Soochow University, Changzhou, People's Republic of China
| | - Wu Huang
- Department of Neurosurgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, People's Republic of China
| | - Hong Zhu
- Department of Radiation Oncology, Minhang Branch of Cancer Hospital of Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
64
|
Pharmacological inhibition of p38 potentiates antimicrobial peptide TP4-induced cell death in glioblastoma cells. Mol Cell Biochem 2019; 464:1-9. [PMID: 31673920 DOI: 10.1007/s11010-019-03643-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 10/23/2019] [Indexed: 12/24/2022]
Abstract
Glioblastoma is the most common and deadly type of brain cancer. The poor prognosis may be largely attributed to inadequate disease response to current chemotherapeutic agents. Activation of p38 is associated with deleterious outcomes in glioblastoma patients, as its signaling mediates chemoresistance mechanisms. Antimicrobial peptide tilapia piscidin (TP) 4 was identified from Nile tilapia (Oreochromis niloticus) and exhibits strong bactericidal effects on Gram-positive and Gram-negative bacteria. TP4 also has anticancer activity toward human triple-negative breast cancer cells and glioblastoma cells. In the present study, we tested the cytotoxic effects of combined TP4 and p38 inhibitors on glioblastoma U251 cells. We found that the combination of TP4 and p38 inhibitors (SB202190 and VX-745) enhanced cytotoxicity in U251 glioblastoma cells but not noncancerous neural cells. Cytotoxicity from the combination treatments proceeded via necrosis and not apoptosis. Mechanistically, SB202190 potentiated TP4-induced mitochondrial dysfunction, reactive oxygen species generation and unbalanced antioxidant status, which resulted in necrotic cell death. Thus, we demonstrated for the first time that combinations of TP4 and p38 inhibitors have the potential to preferentially target glioblastoma cells, while sparing noncancerous neural cells.
Collapse
|
65
|
Lo Dico A, Salvatore D, Martelli C, Ronchi D, Diceglie C, Lucignani G, Ottobrini L. Intracellular Redox-Balance Involvement in Temozolomide Resistance-Related Molecular Mechanisms in Glioblastoma. Cells 2019; 8:cells8111315. [PMID: 31653091 PMCID: PMC6912456 DOI: 10.3390/cells8111315] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/18/2019] [Accepted: 10/22/2019] [Indexed: 01/01/2023] Open
Abstract
Glioblastoma (GBM) is the most common astrocytic-derived brain tumor in adults, characterized by a poor prognosis mainly due to the resistance to the available therapy. The study of mitochondria-derived oxidative stress, and of the biological events that orbit around it, might help in the comprehension of the molecular mechanisms at the base of GBM responsiveness to Temozolomide (TMZ). Sensitive and resistant GBM cells were used to test the role of mitochondrial ROS release in TMZ-resistance. Chaperone-Mediated Autophagy (CMA) activation in relation to reactive oxygen species (ROS) release has been measured by monitoring the expression of specific genes. Treatments with H2O2 were used to test their potential in reverting resistance. Fluctuations of cytoplasmic ROS levels were accountable for CMA induction and cytotoxic effects observed in TMZ sensitive cells after treatment. On the other hand, in resistant cells, TMZ failed in producing an increase in cytoplasmic ROS levels and CMA activation, preventing GBM cell toxicity. By increasing oxidative stress, CMA activation was recovered, as also cell cytotoxicity, especially in combination with TMZ treatment. Herein, for the first time, it is shown the relation between mitochondrial ROS release, CMA activation and TMZ-responsiveness in GBM.
Collapse
Affiliation(s)
- Alessia Lo Dico
- Department of Pathophysiology and Transplantation, University of Milan, 20090 Segrate (MI), Italy.
| | - Daniela Salvatore
- Department of Pathophysiology and Transplantation, University of Milan, 20090 Segrate (MI), Italy.
- Doctorate School of Molecular and Translational Medicine, University of Milan, 20122 Milan, Italy.
| | - Cristina Martelli
- Department of Pathophysiology and Transplantation, University of Milan, 20090 Segrate (MI), Italy.
| | - Dario Ronchi
- Neurology Unit, Neuroscience Section, Department of Pathophysiology and Transplantation, Dino Ferrari Centre, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, 20122 Milan, Italy.
| | - Cecilia Diceglie
- Department of Pathophysiology and Transplantation, University of Milan, 20090 Segrate (MI), Italy.
| | | | - Luisa Ottobrini
- Department of Pathophysiology and Transplantation, University of Milan, 20090 Segrate (MI), Italy.
- Molecular Bioimaging and Physiology (IBFM), CNR, 20090 Segrate (MI), Italy.
| |
Collapse
|
66
|
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: 17] [Impact Index Per Article: 3.4] [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
|
67
|
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: 3.8] [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
|
68
|
ANKRD49 inhibits etoposide-induced intrinsic apoptosis of GC-1 cells by modulating NF-κB signaling. Mol Cell Biochem 2019; 457:21-29. [DOI: 10.1007/s11010-019-03508-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 02/14/2019] [Indexed: 01/09/2023]
|